Abstract
Sphingosine-1phosphate (S1P), platelet activating factor (PAF) and eicosanoids are bioactive lipid mediators abundantly produced by antigen-stimulated mast cells that exert their function mostly through specific cell surface receptors. Although it has long been recognized that some of these bioactive lipids are potent regulators of allergic diseases, their exact contributions to disease pathology have been obscured by the complexity of their mode of action and the regulation of their metabolism. Indeed, the effects of such lipids are usually mediated by multiple receptor subtypes that may differ in their signaling mechanisms and functions. In addition, their actions may be elicited by cell surface receptor-independent mechanisms. Furthermore, these lipids may be converted into metabolites that exhibit different functionalities, adding another layer of complexity to their overall biological responses. In some instances, a second wave of lipid mediator synthesis by both mast cell and non-mast cell sources may occur late during inflammation, bringing about additional roles in the altered environment. New evidence also suggests that bioactive lipids in the local environment can fine-tune mast cell maturation and phenotype, and thus their responsiveness. A better understanding of the subtleties of the spatiotemporal regulation of these lipid mediators, their receptors and functions may aid in the pursuit of pharmacological applications for allergy treatments.
Keywords: mast cells, sphingosine-1-phosphate, eicosanoids, platelet activating factor, lipid mediators, allergy
1- INTRODUCTION
Mast cells differentiate in peripheral tissues from myeloid lineage progenitors. Influenced by stem cell factor (SCF) and other signals in their environment, they acquire their characteristic granularity by storing a variety of substances, including proteases, proteoglycans and vascular mediators in intracellular vesicles (Douaiher et al., 2014; Galli et al., 2005; Metz et al., 2007; Olivera and Rivera, 2014). Concomitantly, they gain cell surface expression of the high affinity receptor for IgE, FcεRI. This allows the mast cell to respond to an antigen by releasing the contents of its granules (degranulation), a critical process for the initiation of the immediate allergic response (Galli and Tsai, 2012; Rivera and Gilfillan, 2006). Antigen-stimulated mast cells also actively produce and secrete a wide variety of lipids and proteins that require de novo synthesis (Blank et al., 2014; Galli et al., 2005; Metz et al., 2007). Among the lipid mediators that mast cells abundantly synthesize are eicosanoids (prostaglandins and leukotrienes), platelet activating factor (PAF) and sphingosine-1-phosphate (S1P) (Boyce, 2007; Mencia-Huerta et al., 1983; Olivera, 2008). These mediators are exported from mast cells within minutes after stimulation (eicosanoids and PAF) or at later times (S1P) and act in the surrounding environment by binding to various types of cognate receptors from the G-protein coupled receptor superfamily (GPCR), which are ubiquitously expressed in tissues and cells. These lipid-binding receptors modulate host defense and the allergic immune response, among other biological processes, by affecting vascular permeability and contractility, chemotaxis of immune cells to sites of inflammation and by inducing varied responses in stromal cells (Boyce, 2007; Honda et al., 2002; Rivera et al., 2008; Serhan et al., 2008). Because most of the above mentioned lipid mediators may bind several types of distinct receptors and each receptor is poised to generate unique downstream signals by virtue of their coupling to varied Gα subunits, the predominant biological function that results may depend on the population of cells present in the tissue as well as the quantitative and qualitative differences in the receptors engaged. Consequently, engagement of specific lipid mediator receptors may mediate pro-inflammatory functions or contribute to the resolution of inflammation depending on the tissue they act on and the timing of action.
Although cell surface expression of FcεRI and KIT (the receptor for SCF) and high metachromatic granularity are common hallmarks of differentiated mast cells, the granule content, life span and functionality of these cells can vary significantly depending on the surrounding microenvironment (Bankova et al., 2014; Douaiher et al., 2014; Galli et al., 2005). This is partly due to the diversity of cell surface receptors expressed by mast cells that makes them susceptible to unique environmental signals in the niche they occupy. Since mast cells are long-lived tissue residents with slow turnover (Padawer, 1974), mast cell-derived mediators may influence the differentiation of mast cell progenitors as well as the phenotype of mature mast cells throughout the course of an immune response. For example, it has been recently described that in a mouse model for the “atopic march”, exposure to a given allergen may alter mast cell responses to a different allergen later in life by increasing mast cell numbers and modifying their phenotype from an immuno-suppressive to a pro-inflammatory mast cell (Hershko et al., 2012). Mast cells express a repertoire of lipid mediator receptors, and thus, in addition to their direct contribution to allergic disease (pro- or anti-inflammatory), these lipids may influence mast cell responses and mast cell differentiation or phenotype, altering their potential involvement in inflammatory processes. Here we will summarize current knowledge about the production of lipid mediators in mast cells, particularly S1P, and the different aspects of their contribution to allergy.
2- SPHINGOSINE-1-PHOSPHATE (S1P)
Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid metabolite derived from sphingosine, an 18-carbon amino alcohol. Structurally, sphingosine linked to a long fatty acid (ceramide) is the fundamental building block of complex sphingolipids (Hannun and Obeid, 2008). Numerous stimuli can release sphingosine from membrane ceramides, a process catalyzed by cellular ceramidases, and activate one or both sphingosine kinase isoforms (SphK1 and SphK2) that phosphorylate sphingosine to generate S1P intracellularly (Fig. 1) (Olivera, 2008; Taha et al., 2006). S1P thus produced can modulate cellular processes, including those important for inflammation and immune responses, mostly through its G-protein coupled receptors (S1PR1-5) (Hannun and Obeid, 2008; Sanchez and Hla, 2004), a process that requires S1P export from cells by specific transporters. The difference in expression patterns of the five S1P receptors on different cells types confers signaling specificity elicited by the ubiquitous S1P molecule. In addition to its autocrine and/or paracrine mode of action, S1P may also act intracellularly by binding intracellular receptors or targets, adding another layer of complexity and regulation to S1P-mediated signaling (Maceyka and Spiegel, 2014; Olivera, 2008).
Fig. 1. Schematic representation of the biosynthesis of mast cell-associated lipid mediators.
A) Sphingosine-1-phosphate (S1P) is a metabolite of complex sphingolipids. The phosphocholine group of sphingomyelin is cleaved by sphingomyelinase to generate ceramide. The fatty acid in the ceramide moiety is then cleaved by ceramidases to produce sphingosine. Sphingosine can be phosphorylated at the primary hydroxyl group by one of two sphingosine kinases (SphK1 or 2) to form S1P. S1P is terminally degraded by S1P lyase or dephosphorylated back to sphingosine by S1P phosphatases. B) Synthesis of eicosanoids. Cytosolic phospholipase A2 (cPLA2) is translocated to the nuclear membrane to release arachidonic acid (AA) from the sn-2 position of nuclear membrane phospholipids. AA is oxidatively metabolized by cyclooxygenase (COX) and lipoxygenase (5-LO) producing the precursors for prostaglandins (PG) and leukotrienes (LT), respectively. There are two COX isoenzymes: COX-1 (constitutive) and COX-2 (inducible). Either one catalyze sequential reactions: first, 2 molecules of oxygen are inserted into AA, cyclizing it to produce PGG2; next, an endoperoxidase reaction reduces PGG2 to PGH2. PGH2 is a precursor for PG, prostacyclins and tromboxanes (the later two are not depicted). PGH2 is converted into PGD2 by PGD synthase (PGDS) or into PGE2 by PGE synthase (PGES). For the synthesis of LT, 5-LO is translocated to the nuclear envelope oxidizing AA to 5-hydroxyperoxyeicosatetraenoic acid (5-HPETE) and then dehydrating it to leukotriene A4 (LTA4). The generation of LTA4 by 5-LO also depends on the function of 5-LO activating protein (FLAP), a perinuclear membrane protein that transfers free AA to 5-LO. Two different classes of LTs can be produced from LTA4: LTB4, formed by LTA4 hydrolase (LTA4H), and LTC4, the parent cysteinyl leukotriene (cys-LT), formed by leukotriene C4 synthase (LTC4S). LTC4 is then exported from the cell, where it is further converted to LTD4 and LTE4, the most stable cys-LT. C) For the synthesis of platelet-activating factor (PAF), a fatty acid is removed from the sn-2 position of ether-linked phospholipids (1-alkyl-2-acyl-sn-glycero-3-phosphorylcholine) by phospholipase A2 (PLA2) to produce the intermediate lyso-PAF (1-alkyl-sn-glycero-3-phosphorylcholine). An acetyl group is then transferred to lyso-PAF by lyso-PAF acetyltransferase (LPCAT) to produce PAF. Deacetylation of PAF by PAF acetylhydrolase (PAF-AH) inactivates PAF.
A critical aspect for S1P biological functions is that, under homeostatic conditions, there is a sharp differential between the levels of S1P in circulation (micromolar range) and those found in tissues (nanomolar range) (Olivera et al., 2013a; Schwab et al., 2005). The high S1P concentrations in blood and lymph signal immune cells to migrate out of primary and secondary lymphoid organs (low S1P zones) into circulation (high S1P zones). In addition, the presence of S1P micro-gradients within the tissue can regulate movement of immune cells to and from particular substructures (Arnon and Cyster, 2014; Moriyama et al., 2014). The steep S1P gradient between blood and organs is maintained by enzymatic activities that constantly degrade S1P in tissues (mainly S1P lyase (Schwab et al., 2005; Serra and Saba, 2010) and two specific S1P phosphatases (SPP1 and SPP2) (Olivera et al., 2013a)) (Fig. 1). On the other hand, the major sources of circulating S1P are red blood cells and endothelial cells (Olivera et al., 2013a; Pappu et al., 2007). Aberrant maintenance of S1P gradients can cause lymphopenia, vascular barrier dysfunction, and other abnormalities.
Mast cells, through engagement of FcεRI, KIT or IL-3 receptors (and possibly other receptors) generate and release abundant quantities of S1P. This may cause temporary increases in the levels of S1P in tissues that can effectively engage S1P receptors in surrounding cells and elicit responses (Choi et al., 1996; Jolly et al., 2004; Olivera et al., 2006; Prieschl et al., 1999). Furthermore, mast cell-generated S1P controls critical mast cell effector functions. Genetic deletion or silencing of SphKs results in pronounced deficiencies in both immediate and delayed mast cell responses, including degranulation, cytokine and eicosanoid production and chemotaxis (Dillahunt et al., 2013; Mitra et al., 2006; Olivera et al., 2007). Both isoforms of sphingosine kinase (SphK1 and 2) are activated upon antigen stimulation (Fig. 2) (Dillahunt et al., 2013; Olivera et al., 2006; Oskeritzian et al., 2008). However, while SphK1 is key for regulating most human mast cell responses, SphK2 is predominant in most mouse mast cell responses. In addition to the species of origin, the role or predominance of each isoform in particular mast cell functions seems to depend on the tissue of origin and the stage of mast cell differentiation (Dillahunt et al., 2013). We will briefly review the mechanisms of S1P production and release following engagement of FcεRI, and the current view of how mast cell-generated S1P may affect both, mast cell phenotype and function as well as the surrounding microenvironment.
Fig. 2. Bioactive lipid mediators and their function in mast cell biology.
Eicosanoids (leukotrienes (LT) and prostaglandin D2 (PGD2)) as well as platelet-activating factor (PAF) are generated and released by mast cells within minutes of mast cell activation, while the release of sphingosine1-phosphate (S1P) peaks hours after activation. FcεRI induces the activation of phospholipase A2 (PLA2) and sequentially other enzymes depicted in Fig. 1, leading to the biosynthesis LT, PGD2 and PAF. FcεRI aggregation also causes the early activation of sphingosine kinases (SphK) to generate S1P. These lipids are released by specific transporters (not depicted) and engage their specific GPCR in both autocrine and paracrine manners. Activation of these receptors in mast cells by mast cell generated lipid mediators can modify responses such as degranulation, cytokine production, chemotaxis and proliferation. S1P may also mediate some of these responses independently of its cell surface receptors, acting as an intracellular signaling molecule. Mast cell-expressed GPCR receptors can also be activated in a paracrine manner by lipid mediators released by other cell types, as has been described for the PGD2 receptor DP1. The soluble form of group III PLA2 (PLA2G3) secreted from mast cells increases the expression of fibroblastic, lipocalin-type PGD2 synthase (L-PGDS). PGD2 produced by fibroblasts, but not by mast cells, engages mast cell DP1 receptors and promotes mast cell maturation. For clarity, the distinct receptors and functions for each particular class of lipid mediators are not depicted separately. For their detailed function, see Table I and the text.
2.1- S1P generation and export by IgE/antigen stimulated mast cells
Upon IgE/antigen-mediated activation of mast cells, FcεRI receptors associate with lipid rafts where sphingolipids are enriched. Activation of FcεRI facilitates the recruitment of both SphK1 and SphK2 to lipid rafts in proximity to their substrate via the Src kinase family members Fyn and, particularly, Lyn (Olivera et al., 2006; Urtz et al., 2004). Fyn and Lyn also provide additional signals for the activation of SphKs. For example, downstream of Fyn, the adaptor protein GRB2-associated-binding protein 2 (Gab2) and PI3K activity are required for maximal activation of SphKs (Olivera et al., 2006). While Fyn deficiency results in a complete loss of SphK activation, Lyn deficiency delays the activation of SphKs (Urtz et al., 2004). As a consequence of Fyn and Lyn activities, a peak in intracellular S1P levels ensues within minutes of FcεRI engagement (Olivera et al., 2006) but this early rise is not accompanied by a detectable release into the extracellular space (Mitra et al., 2006; Olivera, 2008) and it may be linked to its regulation of mast cell responses. A second phase in the production of S1P occurs after 30 min, coinciding with the maximal activity of SphK (Olivera et al., 2006), and precedes its transport out of mast cells and appearance in the extracellular media. Although S1P in the extracellular space has also been detected in low amounts at earlier time points, this was observed after pre-loading mast cells with [3H]-Sphingosine (Mitra et al., 2006), which itself may alter this response.
Efficient transport of charged S1P molecules across cellular membranes requires active export by specific transporters. A recently identified bona-fide transporter for S1P is a member of the major facilitator superfamily (MFS), Spns2. Its role in S1P export has been demonstrated in zebrafish and in mice with a major impact on embryogenesis and lymphocyte trafficking, respectively, processes where extracellular S1P plays important roles (Fukuhara et al., 2012; Kawahara et al., 2009). Members of the ATP binding cassette (ABC) superfamily of transporters have also been implicated in S1P transport (Maceyka and Spiegel, 2014). In activated mast cells, knockdown or pharmacological inhibition of ABCC1 significantly hinders FcεRI-dependent export of S1P (Mitra et al., 2006). It remains to be determined whether Spns2 or other ABC transporters play a contributory role in the export of S1P from mast cells.
2.2- Effect of S1P on mast cells
2.2.1- S1PR expression and regulation of mast cell function
Mast cells have been reported to express two out of the five receptors for S1P: S1P1 and S1P2 receptors. S1P generated following FcεRI-mediated SphK activation was shown to induce ligand-dependent transactivation of both S1P1 and S1P2 (Jolly et al., 2004). Accumulated data from studies examining the role of S1P receptors on mast cell function support a model where SphK activation and S1P1 signaling are involved in the migration of mast cells toward antigen (Jolly et al., 2004; Mitra et al., 2006) (Fig. 2). Accordantly, in a model of food allergy, SphK-deficient mast cells failed to migrate to the gastric mucosa in response to antigen challenges (Diesner et al., 2012). Pharmacological inhibition, genetic deletion or knockdown of SphK or S1P1 receptor in mast cells inhibited chemotactic motility towards antigen or SCF without affecting chemotaxis towards S1P, suggesting an autocrine function for antigen or SCF-generated S1P (Dillahunt et al., 2013; Jolly et al., 2004; Olivera et al., 2006). In support for this concept, inhibition of ABCC1- mediated S1P export blocks migration of mast cells to antigen in vitro, an effect that is rescued by addition of S1P (Mitra et al., 2006).
S1P2 receptor, on the other hand, has been described to enhance FcεRI-induced degranulation (Jolly et al., 2004; Mitra et al., 2006; Olivera et al., 2006) (Fig. 2). However, this effect may depend on the type of mast cell and is likely influenced by environmental cues that may affect S1P2 receptor expression (Jolly et al., 2004; Olivera, 2008). Our data using S1P2-deficient mast cells did not support a role for S1P2 receptors in the degranulation of antigen-stimulated peritoneal or bone marrow derived mast cells (BMMC) differentiated in the presence of SCF and IL-3 in the culture media, and mice deficient in S1P2 did not show alterations in antigen-induced immediate hypersensitivity reactions (Olivera et al., 2013b). However, a role for S1P2 in FcεR-induced degranulation was observed in BMMC differentiated only with IL-3 and in RBL-2H3 cells (Jolly et al., 2004; Olivera et al., 2013b). In human mast cells, the effect of silencing S1P2 on degranulation varied from markedly pronounced (Mitra et al., 2006; Oskeritzian et al., 2010) to very minimal (Olivera et al., 2013b). Furthermore, when exogenous S1P alone or in combination with antigen is added to mast cells the reported effect on degranulation also varies from pronounced (Oskeritzian et al., 2010) to very marginal (Bansal et al., 2008; Jolly et al., 2004; Olivera et al., 2006). The mechanism by which S1P2 might regulate mast cell degranulation still remains unclear. S1P does not appear to be released into the media before the occurrence of degranulation, and inhibition of ABCC1-mediated S1P export does not affect FcεRI-induced degranulation (Mitra et al., 2006). Thus, an autocrine “inside-out” model of S1P signaling is questionable, unless another type of transporter couples intracellular S1P to the cell membrane receptor. Alternatively, S1P translocated to the outer leaflet of the plasma membrane could, without being exported, enter by lateral diffusion into the binding pocket of the receptor, a potential path that was evidenced in crystallography studies of S1P1 receptor (Hanson et al., 2012). S1P1 has been recently shown to be present within intracellular multivesicular endosomes where it may support exosome maturation and release (Kajimoto et al., 2013). If a comparable mechanism exists for S1P2 in mast cells, S1P generated by antigen stimulation may not require export to mediate degranulation in certain populations of mast cells. However, these possibilities remain theoretical.
2.2.2-S1P receptor-independent effects on mast cell function
The biological effects of S1P, beyond those conferred by activation of S1P receptors, may be mediated through its binding to intracellular targets. Although not many intracellular S1P targets have been identified, it was reported that S1P interacts with TNF receptor-associated factor 2 (TRAF2) resulting in the activation of TRAF2 E3 ligase activity and modulation of NF-κB signaling (Alvarez et al., 2010). S1P can also modulate gene transcription by directly binding to histone deacetylases and inhibiting their activity (Hait et al., 2009).
Mouse mast cells lacking SphK2, which do not produce or export S1P, have a profound defect in calcium mobilization, degranulation, eicosanoid and cytokine production in response to antigen and these defects are either not restored (calcium) or partially restored (degranulation) when S1P is added exogenously (Olivera et al., 2007). This may suggest, with the exception of the considerations pointed in the section above, that S1P receptors are not major contributors to degranulation and that S1P might modulate mast cell function as an intracellular mediator (Fig. 2) or by affecting the relative levels of other lipid products such as SPH and ceramide whose effects generally oppose those of S1P (Prieschl et al., 1999). However, the demonstration that S1P effects on early mast cell responses are linked to its direct binding to intracellular targets or that are receptor-independent (by using mast cells deficient in multiple S1P receptors) is still lacking.
2.2.3- Effect of S1P on mast cell phenotype
In addition to the role of mast cell-generated S1P on mast cell responses upon antigen stimulation, other studies suggest that S1P may act as an environmental cue that shapes the phenotype of mast cells. This was hypothesized after the observation that mast cells deficient in SphK2, which produce very little S1P and are unresponsive to allergic challenge in vitro, were paradoxically able to produce normal histamine responses during systemic anaphylaxis in SphK2-deficient mice. These mice exhibit elevated levels of S1P in circulation, and this might compensate for the mast cell-intrinsic inability to produce S1P during stimulation (Olivera et al., 2007). Supporting this hypothesis, the levels of S1P in different mouse strains (C57BL/6 and 129Sv) correlated with the severity of their anaphylactic responses, with the TH1-dominated C57BL/6 mice exhibiting lower levels of S1P and anaphylactic responses (Olivera et al., 2013c) than the TH2-dominated 129Sv strain. However, when the levels of S1P in C57BL/6 were increased by adding an inhibitor of S1P lyase in the drinking water for days before challenge, mice showed enhanced systemic anaphylactic responses similar to those in 129Sv mice, suggesting a link between S1P levels and the enhanced mast cell phenotype (Olivera et al., 2013c). In agreement, chronic exposure to S1P during differentiation of BMMC cultures resulted in a hyper-responsive phenotype that was maintained for at least two weeks after suspension of treatment and, when engrafted into mast cell-deficient KitW-sh/W-sh mice, produced stronger anaphylactic responses than untreated cells. Furthermore, defective catabolism of S1P due to S1P-lyase deficiency in BMMC resulted in a hyperresponsive phenotype and an altered gene expression landscape, supporting the idea that chronic exposure of mast cells to elevated S1P causes phenotypic changes (Olivera et al., 2013c). The specific involvement of S1P receptors in facilitating these phenotypical changes is currently unknown.
Studies in human cord blood progenitors demonstrated that chronic exposure to both S1P and SCF enhances the rate of differentiation into mast cells as well as the number of connective tissue-type mast cells (Price et al., 2009). The mechanism for how S1P induces a developmental shift in the phenotype of human mast cells appears to be partially indirect, involving the production of IL-6 by cells of the monocyte/macrophage lineage (Price et al., 2009). Altogether, these studies suggest that deregulation of S1P homeostasis in circulation or in the tissue environment by diet, genetic or environmental factors, may result in alterations in mast cell phenotype and the susceptibility to particular allergic reactions.
2.3- Effect of S1P on the allergic response
As discussed earlier, under homeostatic conditions, the levels of interstitial S1P in tissues are markedly low compared to the levels of S1P in circulation (Olivera et al., 2013a). However, changes in the tissue environment can result in local increases in S1P concentrations. For instance, in various models of acute inflammation (Keul et al., 2011; Ledgerwood et al., 2008; Roviezzo et al., 2011), S1P levels were found to be elevated in tissue exudates. Similarly, in asthma patients, S1P is elevated in the bronchoalveolar lavage hours after an allergic challenge, suggesting local changes in S1P (Ammit et al., 2001). Mast cells respond to environmental stimuli and can synthesize and release significant quantities of S1P into the media and thus have the potential to alter interstitial S1P concentrations. Because mast cells rapidly degranulate and release vasoactive mediators, S1P levels in the interstitium may immediately increase due to plasma leakage and flooding of blood-born S1P. Active release of S1P from mast cells, which occurs at later times following stimulation, may further increase S1P levels and significantly modulate inflammatory processes. Given that S1P is an important regulator of angiogenesis, vascular permeability (Blaho and Hla, 2014) and immune cell recruitment/function (Rivera et al., 2008), the presence of mast cell-derived S1P likely contributes significantly to the regulation of allergic inflammation. Little is known, however, about the specific contributions of increased S1P on the allergic response, and both pro- and anti-inflammatory roles have been proposed. This is due, in part, to the complex nature of S1P, including the ubiquitousness of its synthesis and receptor expression, the pleiotropic biological functions of S1P, and the possible conversion of S1P to sphingosine or ceramide, which mediate distinct biological functions. The specific function of S1P during inflammation may depend on the nature of a particular antigenic challenge, the type of immune cells that respond to this challenge, and the site of action for S1P and whether the tissue has changed due to chronic inflammatory insults.
2.3.1-Pro-inflammatory roles of S1P
During the early stages of inflammation, S1P may promote inflammation by attracting diverse immune cells to the site (Arnon and Cyster, 2014; Rivera et al., 2008; Spiegel and Milstien, 2011) or by retaining hematopoietic stem cell progenitors (Massberg et al., 2007) and mature T cells in tissues through increased adhesion to the afferent lymphatic endothelium (Ledgerwood et al., 2008). Several reports suggest that S1P can also affect the nature of immune responses by polarizing CD4+ T cells into different subsets. While some studies find that S1P skews T cell responses toward allergic TH2-phenotypes and disfavors TH1-cell responses, either directly or partly mediated by its effects on DCs (Reviewed in (Rivera et al., 2008), others indicate that S1P, via the S1P1 receptor, suppresses the differentiation of both thymic and extrathymic T-reg cells and promotes TH1-cell differentiation (Liu et al., 2010). In addition, S1P has also been reported to promote TH17 differentiation via T cell-expressed S1P1 receptor (Garris et al., 2013; Huang et al., 2007) or indirectly, via DC-expressed S1P4 receptor (Schulze et al., 2011). The exact conditions that define these different outcomes are not entirely clear. In aggregate, the notion remains that S1P may serve as a pro-inflammatory signal by promoting T cell effector differentiation and by suppressing T-regs, a T cell subset that also modulates antigen-induced responses in mast cells (Gri et al., 2008). This may be particularly relevant in cases of recurrent allergic reactions such as those seen with allergic asthma (Galli and Tsai, 2012), where repeated increases in S1P (and other mediators) produced by mast cells can thus contribute to chronic inflammation. In addition, the repeated challenges inherent to chronic allergic diseases cause tissue remodeling characterized by epithelial cell injury and structural changes in vascular, smooth muscle and connective tissues, many of which have been shown to be affected by S1P (reviewed in (Ryan and Spiegel, 2008)).
2.3.2- Anti-inflammatory roles of S1P
Other lines of evidence suggest, however, an anti-inflammatory role for S1P. Skin biopsies of individuals with atopic dermatitis (Seo et al., 2006) and psoriasis (Mechtcheriakova et al., 2007) show increased expression of two enzymes involved in the degradation of S1P, S1P Lyase and S1P-phosphatase type 2 (SPP2), respectively (Fig. 1). In agreement, the degradation of S1P was increased and S1P levels reduced in lesional skin of dogs with atopic dermatitis compared to normal skin (Baumer et al., 2011). A possible interpretation is that low S1P levels allow for perpetuation of the disease, which infers a protective role for S1P. Topical application of S1P to the ear in mouse models of dermatitis or psoriasis reduced ear swelling and hyperproliferation of the skin (Reines et al., 2009; Schaper et al., 2013). Furthermore, inhalation of S1P or the S1P mimetic FTY720 suppressed airway inflammation and bronchial hyperresponsiveness in a model of allergic asthma (Idzko et al., 2006). In both models, the mechanism for such suppression involved prevention of dendritic or Langerhans cell migration to the lymph nodes (Idzko et al., 2006; Reines et al., 2009) and in the dermatitis model, reduced antigen processing via S1P2 receptor activation (Japtok et al., 2012). S1P has also been reported to exert anti-inflammatory roles by promoting the switch from the pro-inflammatory M1 to the anti-inflammatory M2 macrophage subtype (reviewed in (Rivera et al., 2008)).
2.3.3- A protective role for non-mast cell derived S1P in anaphylaxis
S1P production during systemic anaphylaxis, a severe and sometimes fatal allergic reaction affecting multiple organs, may help reduce the severity of anaphylactic shock (Olivera et al., 2010). Even though the cellular source for protective S1P during systemic anaphylaxis is not the mast cell and is non-hematopoietic (Olivera et al., 2010), it will be briefly discussed here because of the important implications for anaphylaxis.
In models of passive IgE/antigen- or histamine-induced anaphylaxis (Olivera et al., 2013b; Olivera et al., 2010) S1P is produced via SphK1 (probably in the vascular beds) and facilitates recovery from anaphylaxis via the activation of S1P2, which counteracts vasodilation and hypotension. In more severe models of anaphylaxis, such as those induced by PAF (Camerer et al., 2009; Cui et al., 2013), IgE clone SPE-7 (Camerer et al., 2009; Olivera et al., 2013b) and active systemic anaphylaxis (Cui et al., 2013), S1P1 (Camerer et al., 2009) and S1P2 (Cui et al., 2013) were shown to be protective by regulating inter-endothelial cell gaps and vascular permeability. S1P2 suppressed endothelial nitric oxide synthase (eNOS) stimulation and NO production associated with anaphylaxis, thus controlling the disassembly of adherens junctions in the endothelium and excessive permeability (Cui et al., 2013). Thus, while it appears that the generation of S1P during anaphylaxis and/or the presence of S1P in circulation may be of importance for the recovery from shock, the dominant involvement of S1P1 and S1P2 in maintaining vascular tone or vascular barrier integrity may depend on the strength or type of stimuli, being their role on vascular integrity revealed under circumstances where vascular leakage is most severe. Although the function of S1P receptors in human anaphylaxis has not yet been explored, based on the mouse models and the beneficial effect of S1P injection in histamine-induced anaphylaxis (Olivera et al., 2010), it is tempting to speculate that a combination of S1P1 and S1P2 agonists could be an option for alternative anaphylaxis treatments. Other studies, however, have pointed to a role for S1P2 in promoting endothelial cell permeability and inflammatory responses of the endothelium during endotoxemia (Sanchez et al., 2007; Zhang et al., 2013) and thus, it still remains to be understood how the same receptor can counteract PAF-induced vasopermeability while promoting permeability by other inflammatory insults.
3- EICOSANOIDS
Eicosanoids are biologically active lipids derived from the oxidation of 20-carbon fatty acids, particularly the polyunsaturated and essential fatty acid arachidonic acid (AA), and include prostaglandins, thromboxanes, leukotrienes and other oxygenated derivatives. By the early 1980s, it was recognized that activated mast cells could rapidly release abundant quantities of AA and subsequently metabolize it to prostaglandin D2 (PGD2) and cysteinyl leukotrienes (Cys-LT) (also known as slow reacting substance of anaphylaxis) with potent inflammatory effects in allergic diseases (Lewis and Austen, 1981). Consequent studies revealed that although mast cells can generate leukotriene B4 (LTB4), the cysteinyl leukotriene C4 (LTC4) and PGD2 upon activation, the specific profile or relative amounts of eicosanoid generation varies in both human and rodent mast cells depending on the tissue of origin, the stage of differentiation and culture conditions (Lundstrom et al., 2013) and reviewed in (Boyce, 2007). PGE2 has not been considered to be an eicosanoid abundantly produced by antigen-stimulated mast cells in culture (Boyce, 2007) although a recent report detected higher intracellular content of PGE2 than other prostanoids by mass spectrometry in mast cells isolated from inflamed tissue in a delayed type hypersensitive model (Thomas et al., 2014). Even though the release of eicosanoids by antigen-activated mast cells requires several enzymatic steps for their de novo production (Fig. 1) and transporter-mediated export (Fletcher et al., 2010), it occurs rapidly (within 10-30 minutes), contributing to early onset allergic responses.
3.1- Prostaglandins
3.1.1- Prostaglandin D2
PGD2 production is mediated by the sequential enzymatic activities of cyclooxygenase (COX) and prostaglandin D2 synthase (PGDS) (Murakami et al., 1994) (Fig. 1). Although other cells also produce PGD2, mast cells may be a predominant source of PGD2 (and its metabolites) in peripheral tissues affected by allergic pathologies such as nasal polyposis (Nantel et al., 2004), eosinophilic chronic rhinosinusitis with nasal polyps (Cao et al., 2014) and eosinophilic esophagitis (Abonia et al., 2010). Indeed, PGD2 has been proposed as a selective indicator of mast cell activation in some diseases such as bronchial asthma and mastocytosis (Bochenek et al., 2004; Morrow et al., 1995). The transcription of enzymes involved in PGD2 synthesis in mast cells is enhanced by inflammatory and environmental signals, and this can augment the PGD2 response to antigen stimulation (Diaz et al., 2006; Murakami et al., 1994; Murakami et al., 1995). Mast cells (as well as macrophages, dendritic cells and TH2 cells) express the hematopoietic isoform of PGDS (H-PGDS) and two isoforms of COX, constitutive (COX-1) and inducible (COX-2). COX-1 is involved in the immediate production of eicosanoids, while COX-2 has been implicated in a delayed phase of PGD2 synthesis (Diaz et al., 2006; Murakami et al., 1994), which appears to be associated with the resolution of inflammation (Scher and Pillinger, 2009).
3.1.1.1- Effect of PGD2 on mast cells
PGD2 actions are mostly mediated by two GPCRs receptors, DP1 and DP2 (also known as chemoattractant receptor-homologous molecule expressed on TH2 cells (CRTH2)) (Boyce, 2007) (Table I). While DP2 is expressed in mast cells, its location in human mast cells was reported to be mostly intracellular and thus they were unresponsive to DP2 agonists alone or in combination with IgE/Ag (Moon et al., 2014). In a mouse model of IgE/Ag-dependent passive cutaneous anaphylaxis, deficiency in DP2 caused increased edema (Taketomi et al., 2013). This may suggest a protective effect of DP2 receptors against vascular leakage or an inhibitory role of this receptor on mast cell degranulation in vivo, which contrasts with the finding that DP2 receptor activation enhances FcεRI-mediated histamine release in basophils (Yoshimura-Uchiyama et al., 2004). Thus, the function of DP2 receptors in mast cells and the mechanism regulating its intracellular location are still unclear.
TABLE I. Lipid mediator receptors most abundantly expressed on mast cells, their: functions on mast cells and on the allergic response.
| Mast cell- expressed GPCR |
Lipid Agonist |
Gα coupled to | Effect on Mast Cell Biology |
Effects on Allergic Responses |
|---|---|---|---|---|
| SIP1 | S1P | Gi | Chemotaxis (+) | Vascular permeability (−) Angiogenesis (+), endothelial cell junctions (+) Immune cell trafficking (+) |
| SIP2 | S1P | Gq, G12/13 | Degranulation (+) | Vascular permeability (+/−) Vasoconstriction, blood pressure regulation Chemotaxis of immune cells (+/−) |
| DP1 | PGD2 | Gs | Promotes maturation and responsiveness to IgE/Ag |
Suppresses TH1-driving cytokines |
| Epithelial activation Inhibition of dendritic cell migration |
||||
| DP2 (CRTH2) |
PGD2 | Gi | Variable effects on degranulation |
Inflammatory cell recruitment, Activation of TH2 cells |
| CysLT1/2 | Cys-LTs | Gi,Gq - CysLT1 |
Degranulation (+) | Inflammatory cell recruitment |
| Cytokine production (+) |
Vascular permeability (+) | |||
| Proliferation (+) | Bronchoconstriction Immune cell function |
|||
| BLT1/2 | LTB4 | Gi | Chemotaxis (+) | Inflammatory cell recruitment |
| Production of Th2− promoting cytokines |
||||
| PAFR | PAF | Gi, Gq/11 | Chemotaxis (+) | Bronchoconstriction, airway hyperreactivity Vascular permeability (+) Hypotension, cardiovascular dysfunction |
| Degranulation (+) |
This table includes only lipid mediator receptors expressed on mast cells that bind lipids abundantly released by mast cells. Non-classical leukotriene receptors are not included.
A recent elegant study uncovered a role for DP1 receptors on the maturation of mast cells, rendering them more responsive to IgE-mediated biological responses (Taketomi et al., 2013) (Table I and Fig. 2). The pathway leading to the activation of DP1 involved a cross-talk between mast cells and fibroblasts whereby a soluble form of group III PLA2 (PLA2G3) secreted from activated mast cells increases the expression of fibroblastic, lipocalin-type PGD2 synthase (L-PGDS). Fibroblast L-PGDS produces PGD2 and, in a paracrine manner, engages mast cell DP1 receptors, which induce the expression of mast cell proteases, hystidine decarboxylase (that produces histamine) and cell surface FcεRI. Mice deficient in PLA2G3, L-PGDS (expressed by fibroblasts) or DP1, and mast cell–deficient mice reconstituted with PLA2G3-null or DP1-null mast cells develop abnormal mast cell phenotypes and reduced responses to systemic or passive cutaneous anaphylaxis, demonstrating that bioactive lipids such as PGD2 may be important cues in the local environment regulating mast cell maturation and responsiveness. Interestingly, deficiency in mast cell-intrinsic H-PGDS seemed to have slightly enhanced cutaneous anaphylaxis reactions in contrast to the attenuated responses in mice lacking fibroblast-intrinsic L-PGDS, which raises the interesting question of how mast cells discriminate between the different pools of PGD2 (Taketomi et al., 2013), a mechanism that needs further investigation.
3.1.1.2- Effect of PGD2 on the allergic response
PGD2 produced mostly by mast cells during the early phase of an allergic reaction has been considered an essential link between the early and late-phase of inflammation by promoting the recruitment of inflammatory cells (Naclerio et al., 1985). DP1 receptors are expressed in dendritic cells and TH1 cells, but not in significantly in TH2 cells while DP2 receptor is viewed as a specific marker for human TH2 cells (Pettipher, 2008; Xue et al., 2005). Both DP1 and DP2 receptors may support a TH2-type inflammatory profile. Whereas DP2 is the major PGD2 receptor contributing to the recruitment of TH2 cells, eosinophils and basophils and promoting TH2 cytokine production (Hirai et al., 2001; Xue et al., 2005), DP1 supports TH2 polarization by suppressing the production of TH1-driving cytokines such IL-12 in dendritic cells (Faveeuw et al., 2003; Pettipher, 2008).
In addition to immune cell recruitment, infusion of PGD2 in humans causes nasal stuffiness, hypotension, and flushing, suggesting an overall participation for PGD2 in immediate hypersensitivity processes (Boyce, 2007; Pettipher, 2008). The higher expression of PGDS, DP1 and DP2 receptors in the nasal mucosa of patients with allergic rhinitis is also consistent with a role for PGD2 in nasal congestion and inflammatory cell infiltration (Nantel et al., 2004). Indeed, a reduction in rhinitis symptoms has been observed after treatment of perennial rhinitis with ramatroban, an effect that has been attributed to its antagonistic effects on DP2 receptors (Pettipher and Whittaker, 2012). Asthma severity also was found to associate with PDG2 in BAL fluid as well as increased H-PGDS (mostly from mast cells) and DP2 expression levels (Fajt et al., 2013). The use of DP2 antagonists has shown relatively modest but significant results in recent clinical trials in some asthmatic populations (reviewed in (Fanning and Boyce, 2013)). A potential role for PGD2 in aspirin exarcerbated respiratory disease (AERD) was revealed in a subset of patients unable to tolerate desensitization to aspirin. These patients showed an overproduction of PGD2 associated with cutaneous and gastrointestinal symptoms after oral aspirin challenge, while patients successfully desensitized exhibited a sharp decrease in PGD2 production (Cahill et al., 2014).
It is important to note, however, that in chronic allergic diseases or delayed hypersensitivity, the role of PGD2 and its receptors may be more complex, involving not only pro-inflammatory functions but also roles in the resolution of inflammation (Boyce, 2007; Rajakariar et al., 2007; Scher and Pillinger, 2009). In part, the pro-resolution effects of PGD2 may be mediated by its dehydration into prostaglandin J2 (15d-PGJ2), a molecule that binds the intracellular peroxisome proliferator-activated receptor-γ (PPAR-γ), although not all the anti-inflammatory effects of PGJ2 appears to be dependent on PPAR-γ (Scher and Pillinger, 2009; Trivedi et al., 2006).
3.1.2- Prostaglandin E2
Prostaglandin E2 (PGE2) is synthetized in multiple cell types following COX and prostaglandin E synthase (PGES) activation (Fig. 1). When released to the extracellular media, PGE2 can activate and signal through four specific GPCRs in target cells (EP1-4) that vary in sensitivity, signaling mechanisms and susceptibility to desensitization, and thus it is not surprising that PGE2 exerts varied cellular responses and mediates pro- and anti-inflammatory functions (reviewed in (Boyce, 2007; Kalinski, 2012; Scher and Pillinger, 2009; Torres et al., 2014)). Mast cells do not appear to be a relevant source for this eicosanoid and thus, the roles of PGE2 in allergy will not be discussed here, but we will briefly highlight the concept that PGE2, through its actions on mast cells, may modulate allergic inflammation.
For instance, PGE2 inhalation before allergen challenge in asthmatic individuals has been known to prevent allergen-induced airway hyperreactivity and inflammation (Gauvreau et al., 1999; Hartert et al., 2000). In mouse asthma models, the anti-inflammatory function of PGE2 has been associated with the inhibition of lung mast cell activity (reviewed in (Torres et al., 2014)). PGE2 dampens FcεRI-induced mediator release in human lung mast cells and certain mast cell subtypes via the Gαs-coupled EP2 receptors, while it enhances responses in other mast cell types mostly when acting through the Gαi-coupled EP3 receptor (Boyce, 2007; Kay et al., 2013; Serra-Pages et al., 2012; Torres et al., 2014). Indeed, variations in the ratio of EP2/EP3 expression in various subtypes of mast cells appears to be a main determinant of the effect of PGE2 on mast cell function (Serra-Pages et al., 2012). Although the involvement of mast cells in the mitigation of asthma by PGE2 has not been fully demonstrated, the use of specific EP2 agonists, which may evade additional and sometimes unwanted effects of PGE2, is an interesting avenue that needs further exploration in the treatment of mast cell-driven allergic diseases and asthma.
3.2- Leukotrienes
While prostaglandins can be synthesized by most cell types, leukotrienes (LTs) are generated by inflammatory cells such as mast cells, macrophages, and neutrophils. In mast cells, activation of FcεRI receptors induces the activation of the enzyme 5-lipoxygenase (5-LO) to produce leukotriene A4 (LTA4), the precursor of LTB4 as well as LTC4, the parent cyteinyl leukotriene (Cys-LT) (Fig. 1). The biological activity of Cys-LTs is largely mediated through two GPCRs referred to as CysLT1 and CysLT2, while LTB4 signals through BLT1 and BLT2 receptors (Table I). Other leukotriene receptors, however, have also been described (Kanaoka and Boyce, 2014; Tager and Luster, 2003). Leukotrienes have long been implicated in allergies and their important and complex contributions to different processes during the allergic response have been recently summarized in excellent reviews (Boyce, 2007; Kanaoka and Boyce, 2014; Laidlaw and Boyce, 2012). Thus, here we will focus on some aspects of the autocrine functions of leukotrienes on mast cells.
3.2.1- Effect of leukotrienes on mast cells
Human mast cells express both CysLT1 and CysLT2 receptors and respond to Cys-LTs by mobilizing calcium, releasing pro-inflammatory cytokines and chemokines, and mediating proliferation (Mellor et al., 2001; Paruchuri et al., 2008) (Fig. 2). Interestingly, the responses to Cys-LTs (as well as those to IgE/antigen) on human mast cells are greatly enhanced by priming with TH2 cytokines, particularly IL-4 (Hsieh et al., 2001; Mellor et al., 2002), an effect mediated in part by an increase in the transcription of LTC4 synthase (LTC4S) (Hsieh et al., 2001) and in the cell surface expression of CysLT2 receptor (Mellor et al., 2003). The CysLT1 receptor is mostly responsible for the augmented mast cell responses to Cys-LTs or to IgE/antigen after IL-4 priming, although CysLT2 and other putative Cys-LT receptors have been reported to contribute (Mellor et al., 2002; Mellor et al., 2003; Paruchuri et al., 2008). IL-4 is produced during allergic inflammation and regulates mast cell proliferation, TH2 cytokine production and FcεRI receptor expression (Lorentz et al., 2005). Thus, the finding that IL-4 regulates Cys-LT synthesis and function may suggest roles for these lipids in the amplification of IL-4 and antigen-induced responses. This may be particularly relevant during the allergic response.
Indeed, IL-4 induces mast cell proliferation partly through the production of Cys-LT, engagement of CysLT1 receptors and transactivation of the receptor for SCF (Jiang et al., 2006), KIT, which is critical for mast cell differentiation, proliferation and function. Thus, the mitogenic pathways induced by CysLT1 receptors are in part convergent with those of KIT, but also distinct (Jiang et al., 2006), as supported by the synergistic effects of Cys-LT and SCF on the proliferation of mast cells (Al-Azzam et al., 2014). On the other hand, SCF can also induce Cys-LT production in mast cells and blockage of its production or antagonism of CysLT1 receptor in vivo, substantially reduces the airway hyperresponsiveness induced after intra-tracheal instillation of SCF in mice (Oliveira et al., 2001). Thus, Cys-LTs can be at the crossroads of FcεRI, IL-4 receptor and KIT pathways, and the regulation, crosstalk and synergy between these pathways may contribute to mast cell hyperplasia and hyper-reactivity during allergic inflammation, a condition where all these regulators are known to be important. This notion is supported by the findings of attenuated airway inflammation and TH2 cytokine production as well as reduced hyperplasia and activation of tracheal intraepithelial mast cells in LTC4S-deficient mice subjected to antigen-induced pulmonary inflammation (Kim et al., 2006).
Mast cells express also the LTB4 receptors, BLT1 and BLT2 (Table I). Both receptors were shown to induce chemotaxis in response to LTB4 (Lundeen et al., 2006), although this effect is particularly prominent in immature mast cells, where the expression of BLT1 is increased compared to mature mast cells (Weller et al., 2005). Indeed, intradermal injection of LTB4 in vivo induced the recruitment of mast cell progenitors to sites of inflammation (Weller et al., 2005). In addition, the BLT2 receptor may mediate, in an autocrine manner, the production of TH2 cytokines by mast cells after FcεRI activation (Cho et al., 2010) (Fig. 2). LTB4 released by mast cells during an allergic trigger may contribute to the recruitment of inflammatory cells as well as circulating mast cell progenitors, perpetuating inflammation.
4- PLATELET-ACTIVATING FACTOR (PAF)
Platelet-activating factor (PAF) is a lipid mediator with an important role in allergy, asthma, as well as other pathological processes, acting through its receptor (PAFR) (Prescott et al., 2000; Tsoupras et al., 2009) (Table I). PAF was originally described as a substance released by basophils in an IgE-dependent process, capable of inducing platelet aggregation. PAF generation by mast cells, basophils and other blood cells requires the activity of PLA2 and a lyso PAF acetyltransferase (Prescott et al., 2000) (Fig. 1). PAF is released during the early phase of the allergic response but has a very short half-life (3 to 13 minutes) due to its inactivation by PAF acetylhydrolase.
4.1- Effects of PAF on mast cells
PAF induces mast cell chemotaxis (Nilsson et al., 2000) and mediator release (Fig. 2). The effects of PAF on mast cell release may vary depending on the mast cell type and the levels of PAF receptor expression. Whereas PAF induced histamine and PGD2 release in mast cells from human lung or derived from adult peripheral blood progenitors, it was ineffective in human skin mast cells in vitro, which do not express the receptor (Kajiwara et al., 2010). However, it can induce degranulation in vivo from skin mast cells when injected intradermally, an effect that may be secondary to its activation of neural reflexes (Petersen et al., 1997). It has been suggested that PAF, released by mast cells or other cells during anaphylaxis may contribute to the dissemination of a mast cell response to an allergen at sites distal to allergen exposure (Kajiwara et al., 2010).
4.2- Effects of PAF on the allergic response
The effects of PAF as a bronchoconstrictor and mediator of airway hyperreactivity in bronchial asthma have long been recognized, although the use of PAF inhibitors as a therapeutic option in asthma has been ineffective (Kasperska-Zajac et al., 2008). PAF also causes severe hypotension and cardiovascular dysfunction during acute anaphylaxis, an effect that, at least in mice, is dependent on a PAF-mediated activation of eNOS and consequent NO formation (Cauwels et al., 2006). In mouse models, PAF antagonists effectively attenuated IgG-induced anaphylaxis (Jiao et al., 2014; Jonsson et al., 2011; Strait et al., 2002; Tsujimura et al., 2008) and peanut-induced anaphylaxis severity (Arias et al., 2009), but did not effectively attenuate IgE-mediated anaphylaxis, which is more dependent on histamine release (Strait et al., 2002). In humans, it has not been clearly established whether anaphylaxis is mediated through a classical IgE-mediated pathway, an alternative IgG-mediated pathway or both (Finkelman, 2007). However, some studies have reported that the levels of PAF in serum correlates with the severity of the anaphylactic reaction while histamine or tryptase levels do not, and PAF acetylhydrolase activity, the enzyme responsible of PAF inactivation, inversely correlates with severity scores (Vadas et al., 2008), and thus PAF antagonists, alone or in combination with other drugs, may be a consideration as rescue therapy for acute anaphylaxis reactions.
5. CONCLUDING REMARKS
Accumulating evidence for the last twenty years have demonstrated that lipid mediators generated by mast cells are critical for the pathogenesis of allergic diseases, yet a full understanding of their intricate and finely tuned participation is incomplete. While mast cell-derived lipid mediators and their receptors represent attractive therapeutic targets for treating allergic inflammation, the pleiotropic functions and complex biology associated with these molecules and the realization of the existence of previously unrecognized receptors mediating some of their actions, pose a significant challenge to researchers who seek to understand and modulate the disease processes they regulate.
To date, several compounds that target lipid mediators have been employed in a clinical setting and the interest in developing and investigating new and more specific compounds is expanding. The S1P receptor modulator, FTY720, was the first FDA-approved sphingo-mimetic oral therapeutic and has been used to treat relapsing-remitting multiple sclerosis (Bigaud et al., 2014), but its local application to treat allergic diseases has been thus far restricted to animal models. Development of more selective agonist and antagonists for S1P receptors or anti-S1P antibodies to locally intercept its actions (Bigaud et al., 2014) may provide interesting therapy options in allergic inflammation and anaphylaxis. While the DP2 receptor antagonist OC000459 has shown promising effects for asthma patients in phase 2 clinical trials, the dual DP1/DP2 antagonist AMG 853 has proven to be less efficacious (reviewed in (Fanning and Boyce, 2013). There are also several specific Cys-LT receptor antagonists (zafirlukast, pranlukast, and montelukast) as well as LT synthesis inhibitors (zileuton) that are in clinical use with variable efficacy (Laidlaw and Boyce, 2012; Montuschi and Peters-Golden, 2010). More recently, a 5-lipoxygenase–activating protein (FLAP) (Fig. 1) inhibitor (GSK2190915) has been shown to be effective in inhibiting allergic responses to inhaled allergen in patients with mild asthma (Kent et al., 2013). The continued development and refinement of compounds that modulate receptors and signaling associated with mast cell-derived lipid mediators will continue to aid in the treatment of human disease and our understanding of the biology these molecules influence.
ACKNOWLEDGMENTS
This work was supported by the Division of Intramural Research Program within NIAID and NIAMS, NIH
ABREVIATIONS
- AA
arachidonic acid
- ABCC1
ATP-Binding Cassette, Sub-Family C
- BLT1-2
leukotriene B4 receptors 1 to 2
- BMMC
bone marrow derived mast cells
- COX
cyclooxygenase
- Cys-LT
cysteinyl leukotrienes
- CysLT1-2
cysteinyl leukotriene receptors 1 to 2
- DP1-2
prostaglandin D2 receptors 1 to 2
- FcεRI
high affinity receptor for IgE
- GPCR
G-protein coupled receptor
- KIT
stem cell factor receptor
- 5-LO
5-lipoxygenase
- LTB4
leukotriene B4
- LTC4
leukotriene C4
- LTC4S
leukotriene C4 synthase
- eNOS
endothelial nitric oxide synthase
- NO
nitric oxide
- PLA2
phospholipase A2
- PAF
platelet activating factor
- PGD2
prostaglandin D2
- PGE2
prostaglandin E2
- PGDS
prostaglandin D synthase
- PI3K
phosphatidylinositol-4,5-bisphosphate 3-kinase
- SphK
sphingosine kinase
- S1P
sphingosine-1-phosphate
- S1P1-5
sphingosine-1-phosphate receptors 1 through 5
- SCF
stem cell factor
- RBL-2H3
basophilic leukemia cell line
- TNF
tumor necrosis factor
- TRAF2
TNF receptor-associated factor 2
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
CHEMICAL COMPOUNDS
FTY720 (CID: 107969)
Leukotriene C4 (CID: 5280493)
Prostaglandin D2 (CID: 448457)
Prostaglandin E2 (CID: 5280360)
Platelet activating factor (CID: 108156)
Sphingosine-1-phosphate (CID: 5283560)
REFERENCES
- Abonia JP, Blanchard C, Butz BB, Rainey HF, Collins MH, Stringer K, Putnam PE, Rothenberg ME. Involvement of mast cells in eosinophilic esophagitis. The Journal of allergy and clinical immunology. 2010;126:140–149. doi: 10.1016/j.jaci.2010.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Al-Azzam N, Kondeti V, Duah E, Gombedza F, Thodeti CK, Paruchuri S. Modulation of mast cell proliferative and inflammatory responses by Leukotriene D and Stem Cell Factor signaling interactions. Journal of cellular physiology. 2014 doi: 10.1002/jcp.24777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alvarez SE, Harikumar KB, Hait NC, Allegood J, Strub GM, Kim EY, Maceyka M, Jiang H, Luo C, Kordula T, Milstien S, Spiegel S. Sphingosine-1-phosphate is a missing cofactor for the E3 ubiquitin ligase TRAF2. Nature. 2010;465:1084–1088. doi: 10.1038/nature09128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ammit AJ, Hastie AT, Edsall LC, Hoffman RK, Amrani Y, Krymskaya VP, Kane SA, Peters SP, Penn RB, Spiegel S, Panettieri RA., Jr. Sphingosine 1-phosphate modulates human airway smooth muscle cell functions that promote inflammation and airway remodeling in asthma. Faseb J. 2001;15:1212–1214. doi: 10.1096/fj.00-0742fje. [DOI] [PubMed] [Google Scholar]
- Arias K, Baig M, Colangelo M, Chu D, Walker T, Goncharova S, Coyle A, Vadas P, Waserman S, Jordana M. Concurrent blockade of platelet-activating factor and histamine prevents life-threatening peanut-induced anaphylactic reactions. The Journal of allergy and clinical immunology. 2009;124:307–314. 314 e301–302. doi: 10.1016/j.jaci.2009.03.012. [DOI] [PubMed] [Google Scholar]
- Arnon TI, Cyster JG. Blood, sphingosine-1-phosphate and lymphocyte migration dynamics in the spleen. Current topics in microbiology and immunology. 2014;378:107–128. doi: 10.1007/978-3-319-05879-5_5. [DOI] [PubMed] [Google Scholar]
- Bankova LG, Dwyer DF, Liu AY, Austen KF, Gurish MF. Maturation of mast cell progenitors to mucosal mast cells during allergic pulmonary inflammation in mice. Mucosal immunology. 2014 doi: 10.1038/mi.2014.91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bansal G, DiVietro JA, Kuehn HS, Rao S, Nocka KH, Gilfillan AM, Druey KM. RGS13 controls g protein-coupled receptor-evoked responses of human mast cells. J Immunol. 2008;181:7882–7890. doi: 10.4049/jimmunol.181.11.7882. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baumer W, Rossbach K, Mischke R, Reines I, Langbein-Detsch I, Luth A, Kleuser B. Decreased concentration and enhanced metabolism of sphingosine-1-phosphate in lesional skin of dogs with atopic dermatitis: disturbed sphingosine-1-phosphate homeostasis in atopic dermatitis. The Journal of investigative dermatology. 2011;131:266–268. doi: 10.1038/jid.2010.252. [DOI] [PubMed] [Google Scholar]
- Bigaud M, Guerini D, Billich A, Bassilana F, Brinkmann V. Second generation S1P pathway modulators: research strategies and clinical developments. Biochimica et biophysica acta. 2014;1841:745–758. doi: 10.1016/j.bbalip.2013.11.001. [DOI] [PubMed] [Google Scholar]
- Blaho VA, Hla T. An update on the biology of sphingosine 1-phosphate receptors. Journal of lipid research. 2014;55:1596–1608. doi: 10.1194/jlr.R046300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Blank U, Madera-Salcedo IK, Danelli L, Claver J, Tiwari N, Sanchez-Miranda E, Vazquez-Victorio G, Ramirez-Valadez KA, Macias-Silva M, Gonzalez-Espinosa C. Vesicular trafficking and signaling for cytokine and chemokine secretion in mast cells. Frontiers in immunology. 2014;5:453. doi: 10.3389/fimmu.2014.00453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bochenek G, Nizankowska E, Gielicz A, Swierczynska M, Szczeklik A. Plasma 9alpha,11beta-PGF2, a PGD2 metabolite, as a sensitive marker of mast cell activation by allergen in bronchial asthma. Thorax. 2004;59:459–464. doi: 10.1136/thx.2003.013573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boyce JA. Mast cells and eicosanoid mediators: a system of reciprocal paracrine and autocrine regulation. Immunological reviews. 2007;217:168–185. doi: 10.1111/j.1600-065X.2007.00512.x. [DOI] [PubMed] [Google Scholar]
- Cahill KN, Bensko JC, Boyce JA, Laidlaw TM. Prostaglandin D: A dominant mediator of aspirin-exacerbated respiratory disease. The Journal of allergy and clinical immunology. 2014 doi: 10.1016/j.jaci.2014.07.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Camerer E, Regard JB, Cornelissen I, Srinivasan Y, Duong DN, Palmer D, Pham TH, Wong JS, Pappu R, Coughlin SR. Sphingosine-1-phosphate in the plasma compartment regulates basal and inflammation-induced vascular leak in mice. The Journal of clinical investigation. 2009;119:1871–1879. doi: 10.1172/JCI38575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cao PP, Zhang YN, Liao B, Ma J, Wang BF, Wang H, Zeng M, Liu WH, Schleimer RP, Liu Z. Increased local IgE production induced by common aeroallergens and phenotypic alteration of mast cells in Chinese eosinophilic, but not non-eosinophilic, chronic rhinosinusitis with nasal polyps. Clinical and experimental allergy: journal of the British Society for Allergy and Clinical Immunology. 2014;44:690–700. doi: 10.1111/cea.12304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cauwels A, Janssen B, Buys E, Sips P, Brouckaert P. Anaphylactic shock depends on PI3K and eNOS-derived NO. The Journal of clinical investigation. 2006;116:2244–2251. doi: 10.1172/JCI25426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cho KJ, Seo JM, Lee MG, Kim JH. BLT2 Is upregulated in allergen-stimulated mast cells and mediates the synthesis of Th2 cytokines. J Immunol. 2010;185:6329–6337. doi: 10.4049/jimmunol.1001213. [DOI] [PubMed] [Google Scholar]
- Choi OH, Kim JH, Kinet JP. Calcium mobilization via sphingosine kinase in signalling by the Fc epsilon RI antigen receptor. Nature. 1996;380:634–636. doi: 10.1038/380634a0. [DOI] [PubMed] [Google Scholar]
- Cui H, Okamoto Y, Yoshioka K, Du W, Takuwa N, Zhang W, Asano M, Shibamoto T, Takuwa Y. Sphingosine-1-phosphate receptor 2 protects against anaphylactic shock through suppression of endothelial nitric oxide synthase in mice. The Journal of allergy and clinical immunology. 2013;132:1205–1214 e1209. doi: 10.1016/j.jaci.2013.07.026. [DOI] [PubMed] [Google Scholar]
- Diaz BL, Satake Y, Kikawada E, Balestrieri B, Arm JP. Group V secretory phospholipase A2 amplifies the induction of cyclooxygenase 2 and delayed prostaglandin D2 generation in mouse bone marrow culture-derived mast cells in a strain-dependent manner. Biochimica et biophysica acta. 2006;1761:1489–1497. doi: 10.1016/j.bbalip.2006.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Diesner SC, Olivera A, Dillahunt S, Schultz C, Watzlawek T, Forster-Waldl E, Pollak A, Jensen-Jarolim E, Untersmayr E, Rivera J. Sphingosine-kinase 1 and 2 contribute to oral sensitization and effector phase in a mouse model of food allergy. Immunology letters. 2012;141:210–219. doi: 10.1016/j.imlet.2011.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dillahunt SE, Sargent JL, Suzuki R, Proia RL, Gilfillan A, Rivera J, Olivera A. Usage of sphingosine kinase isoforms in mast cells is species and/or cell type determined. J Immunol. 2013;190:2058–2067. doi: 10.4049/jimmunol.1201503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Douaiher J, Succar J, Lancerotto L, Gurish MF, Orgill DP, Hamilton MJ, Krilis SA, Stevens RL. Development of mast cells and importance of their tryptase and chymase serine proteases in inflammation and wound healing. Advances in immunology. 2014;122:211–252. doi: 10.1016/B978-0-12-800267-4.00006-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fajt ML, Gelhaus SL, Freeman B, Uvalle CE, Trudeau JB, Holguin F, Wenzel SE. Prostaglandin D(2) pathway upregulation: relation to asthma severity, control, and TH2 inflammation. The Journal of allergy and clinical immunology. 2013;131:1504–1512. doi: 10.1016/j.jaci.2013.01.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fanning LB, Boyce JA. Lipid mediators and allergic diseases. Annals of allergy, asthma & immunology: official publication of the American College of Allergy, Asthma, & Immunology. 2013;111:155–162. doi: 10.1016/j.anai.2013.06.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Faveeuw C, Gosset P, Bureau F, Angeli V, Hirai H, Maruyama T, Narumiya S, Capron M, Trottein F. Prostaglandin D2 inhibits the production of interleukin-12 in murine dendritic cells through multiple signaling pathways. European journal of immunology. 2003;33:889–898. doi: 10.1002/eji.200323330. [DOI] [PubMed] [Google Scholar]
- Finkelman FD. Anaphylaxis: lessons from mouse models. The Journal of allergy and clinical immunology. 2007;120:506–515. doi: 10.1016/j.jaci.2007.07.033. quiz 516-507. [DOI] [PubMed] [Google Scholar]
- Fletcher JI, Haber M, Henderson MJ, Norris MD. ABC transporters in cancer: more than just drug efflux pumps. Nature reviews. Cancer. 2010;10:147–156. doi: 10.1038/nrc2789. [DOI] [PubMed] [Google Scholar]
- Fukuhara S, Simmons S, Kawamura S, Inoue A, Orba Y, Tokudome T, Sunden Y, Arai Y, Moriwaki K, Ishida J, Uemura A, Kiyonari H, Abe T, Fukamizu A, Hirashima M, Sawa H, Aoki J, Ishii M, Mochizuki N. The sphingosine-1-phosphate transporter Spns2 expressed on endothelial cells regulates lymphocyte trafficking in mice. J Clin Invest. 2012;122:1416–1426. doi: 10.1172/JCI60746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Galli SJ, Kalesnikoff J, Grimbaldeston MA, Piliponsky AM, Williams CM, Tsai M. Mast cells as “tunable” effector and immunoregulatory cells: recent advances. Annu Rev Immunol. 2005;23:749–786. doi: 10.1146/annurev.immunol.21.120601.141025. [DOI] [PubMed] [Google Scholar]
- Galli SJ, Tsai M. IgE and mast cells in allergic disease. Nature medicine. 2012;18:693–704. doi: 10.1038/nm.2755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garris CS, Wu L, Acharya S, Arac A, Blaho VA, Huang Y, Moon BS, Axtell RC, Ho PP, Steinberg GK, Lewis DB, Sobel RA, Han DK, Steinman L, Snyder MP, Hla T, Han MH. Defective sphingosine 1-phosphate receptor 1 (S1P1) phosphorylation exacerbates TH17-mediated autoimmune neuroinflammation. Nat Immunol. 2013;14:1166–1172. doi: 10.1038/ni.2730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gauvreau GM, Watson RM, O’Byrne PM. Protective effects of inhaled PGE2 on allergen-induced airway responses and airway inflammation. American journal of respiratory and critical care medicine. 1999;159:31–36. doi: 10.1164/ajrccm.159.1.9804030. [DOI] [PubMed] [Google Scholar]
- Gri G, Piconese S, Frossi B, Manfroi V, Merluzzi S, Tripodo C, Viola A, Odom S, Rivera J, Colombo MP, Pucillo CE. CD4+CD25+ regulatory T cells suppress mast cell degranulation and allergic responses through OX40-OX40L interaction. Immunity. 2008;29:771–781. doi: 10.1016/j.immuni.2008.08.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hait NC, Allegood J, Maceyka M, Strub GM, Harikumar KB, Singh SK, Luo C, Marmorstein R, Kordula T, Milstien S, Spiegel S. Regulation of histone acetylation in the nucleus by sphingosine-1-phosphate. Science. 2009;325:1254–1257. doi: 10.1126/science.1176709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hannun YA, Obeid LM. Principles of bioactive lipid signalling: lessons from sphingolipids. Nat Rev Mol Cell Biol. 2008;9:139–150. doi: 10.1038/nrm2329. [DOI] [PubMed] [Google Scholar]
- Hanson MA, Roth CB, Jo E, Griffith MT, Scott FL, Reinhart G, Desale H, Clemons B, Cahalan SM, Schuerer SC, Sanna MG, Han GW, Kuhn P, Rosen H, Stevens RC. Crystal structure of a lipid G protein-coupled receptor. Science. 2012;335:851–855. doi: 10.1126/science.1215904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hartert TV, Dworski RT, Mellen BG, Oates JA, Murray JJ, Sheller JR. Prostaglandin E(2) decreases allergen-stimulated release of prostaglandin D(2) in airways of subjects with asthma. American journal of respiratory and critical care medicine. 2000;162:637–640. doi: 10.1164/ajrccm.162.2.9904038. [DOI] [PubMed] [Google Scholar]
- Hershko AY, Charles N, Olivera A, Alvarez-Errico D, Rivera J. Cutting edge: persistence of increased mast cell numbers in tissues links dermatitis to enhanced airway disease in a mouse model of atopy. J Immunol. 2012;188:531–535. doi: 10.4049/jimmunol.1102703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hirai H, Tanaka K, Yoshie O, Ogawa K, Kenmotsu K, Takamori Y, Ichimasa M, Sugamura K, Nakamura M, Takano S, Nagata K. Prostaglandin D2 selectively induces chemotaxis in T helper type 2 cells, eosinophils, and basophils via seven-transmembrane receptor CRTH2. The Journal of experimental medicine. 2001;193:255–261. doi: 10.1084/jem.193.2.255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Honda Z, Ishii S, Shimizu T. Platelet-activating factor receptor. Journal of biochemistry. 2002;131:773–779. doi: 10.1093/oxfordjournals.jbchem.a003164. [DOI] [PubMed] [Google Scholar]
- Hsieh FH, Lam BK, Penrose JF, Austen KF, Boyce JA. T helper cell type 2 cytokines coordinately regulate immunoglobulin E-dependent cysteinyl leukotriene production by human cord blood-derived mast cells: profound induction of leukotriene C(4) synthase expression by interleukin 4. J Exp Med. 2001;193:123–133. doi: 10.1084/jem.193.1.123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang MC, Watson SR, Liao JJ, Goetzl EJ. Th17 augmentation in OTII TCR plus T cell-selective type 1 sphingosine 1-phosphate receptor double transgenic mice. J. Immunol. 2007;178:6806–6813. doi: 10.4049/jimmunol.178.11.6806. [DOI] [PubMed] [Google Scholar]
- Idzko M, Hammad H, van Nimwegen M, Kool M, Muller T, Soullie T, Willart MA, Hijdra D, Hoogsteden HC, Lambrecht BN. Local application of FTY720 to the lung abrogates experimental asthma by altering dendritic cell function. The Journal of clinical investigation. 2006;116:2935–2944. doi: 10.1172/JCI28295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Japtok L, Schaper K, Baumer W, Radeke HH, Jeong SK, Kleuser B. Sphingosine 1-phosphate modulates antigen capture by murine Langerhans cells via the S1P2 receptor subtype. PloS one. 2012;7:e49427. doi: 10.1371/journal.pone.0049427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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;177:2755–2759. doi: 10.4049/jimmunol.177.5.2755. [DOI] [PubMed] [Google Scholar]
- Jiao D, Liu Y, Lu X, Liu B, Pan Q, Liu Y, Liu Y, Zhu P, Fu N. Macrophages are the dominant effector cells responsible for IgG-mediated passive systemic anaphylaxis challenged by natural protein antigen in BALB/c and C57BL/6 mice. Cellular immunology. 2014;289:97–105. doi: 10.1016/j.cellimm.2014.03.018. [DOI] [PubMed] [Google Scholar]
- Jolly PS, Bektas M, Olivera A, Gonzalez-Espinosa C, Proia RL, Rivera J, Milstien S, Spiegel S. Transactivation of sphingosine-1-phosphate receptors by FcepsilonRI triggering is required for normal mast cell degranulation and chemotaxis. J Exp Med. 2004;199:959–970. doi: 10.1084/jem.20030680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jonsson F, Mancardi DA, Kita Y, Karasuyama H, Iannascoli B, Van Rooijen N, Shimizu T, Daeron M, Bruhns P. Mouse and human neutrophils induce anaphylaxis. The Journal of clinical investigation. 2011;121:1484–1496. doi: 10.1172/JCI45232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kajimoto T, Okada T, Miya S, Zhang L, Nakamura S. Ongoing activation of sphingosine 1-phosphate receptors mediates maturation of exosomal multivesicular endosomes. Nature communications. 2013;4:2712. doi: 10.1038/ncomms3712. [DOI] [PubMed] [Google Scholar]
- Kajiwara N, Sasaki T, Bradding P, Cruse G, Sagara H, Ohmori K, Saito H, Ra C, Okayama Y. Activation of human mast cells through the platelet-activating factor receptor. The Journal of allergy and clinical immunology. 2010;125:1137–1145 e1136. doi: 10.1016/j.jaci.2010.01.056. [DOI] [PubMed] [Google Scholar]
- Kalinski P. Regulation of immune responses by prostaglandin E2. Journal of immunology. 2012;188:21–28. doi: 10.4049/jimmunol.1101029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kanaoka Y, Boyce JA. Cysteinyl leukotrienes and their receptors; emerging concepts. Allergy, asthma & immunology research. 2014;6:288–295. doi: 10.4168/aair.2014.6.4.288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kasperska-Zajac A, Brzoza Z, Rogala B. Platelet activating factor as a mediator and therapeutic approach in bronchial asthma. Inflammation. 2008;31:112–120. doi: 10.1007/s10753-007-9056-9. [DOI] [PubMed] [Google Scholar]
- Kawahara A, Nishi T, Hisano Y, Fukui H, Yamaguchi A, Mochizuki N. The sphingolipid transporter spns2 functions in migration of zebrafish myocardial precursors. Science. 2009;323:524–527. doi: 10.1126/science.1167449. [DOI] [PubMed] [Google Scholar]
- Kay LJ, Gilbert M, Pullen N, Skerratt S, Farrington J, Seward EP, Peachell PT. Characterization of the EP receptor subtype that mediates the inhibitory effects of prostaglandin E2 on IgE-dependent secretion from human lung mast cells. Clinical and experimental allergy: journal of the British Society for Allergy and Clinical Immunology. 2013;43:741–751. doi: 10.1111/cea.12142. [DOI] [PubMed] [Google Scholar]
- Kent SE, Boyce M, Diamant Z, Singh D, O’Connor BJ, Saggu PS, Norris V. The 5-lipoxygenase-activating protein inhibitor, GSK2190915, attenuates the early and late responses to inhaled allergen in mild asthma. Clin Exp Allergy. 2013;43:177–186. doi: 10.1111/cea.12002. [DOI] [PubMed] [Google Scholar]
- Keul P, Lucke S, von Wnuck Lipinski K, Bode C, Graler M, Heusch G, Levkau B. Sphingosine-1-phosphate receptor 3 promotes recruitment of monocyte/macrophages in inflammation and atherosclerosis. Circulation research. 2011;108:314–323. doi: 10.1161/CIRCRESAHA.110.235028. [DOI] [PubMed] [Google Scholar]
- Kim DC, Hsu FI, Barrett NA, Friend DS, Grenningloh R, Ho IC, Al-Garawi A, Lora JM, Lam BK, Austen KF, Kanaoka Y. Cysteinyl leukotrienes regulate Th2 cell-dependent pulmonary inflammation. J Immunol. 2006;176:4440–4448. doi: 10.4049/jimmunol.176.7.4440. [DOI] [PubMed] [Google Scholar]
- Laidlaw TM, Boyce JA. Cysteinyl leukotriene receptors, old and new; implications for asthma. Clin Exp Allergy. 2012;42:1313–1320. doi: 10.1111/j.1365-2222.2012.03982.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ledgerwood LG, Lal G, Zhang N, Garin A, Esses SJ, Ginhoux F, Merad M, Peche H, Lira SA, Ding Y, Yang Y, He X, Schuchman EH, Allende ML, Ochando JC, Bromberg JS. The sphingosine 1-phosphate receptor 1 causes tissue retention by inhibiting the entry of peripheral tissue T lymphocytes into afferent lymphatics. Nature Immunol. 2008;9:42–53. doi: 10.1038/ni1534. [DOI] [PubMed] [Google Scholar]
- Lewis RA, Austen KF. Mediation of local homeostasis and inflammation by leukotrienes and other mast cell-dependent compounds. Nature. 1981;293:103–108. doi: 10.1038/293103a0. [DOI] [PubMed] [Google Scholar]
- Liu G, Yang K, Burns S, Shrestha S, Chi H. The S1P(1)-mTOR axis directs the reciprocal differentiation of T(H)1 and T(reg) cells. Nat Immunol. 2010;11:1047–1056. doi: 10.1038/ni.1939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lorentz A, Wilke M, Sellge G, Worthmann H, Klempnauer J, Manns MP, Bischoff SC. 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;174:6751–6756. doi: 10.4049/jimmunol.174.11.6751. [DOI] [PubMed] [Google Scholar]
- 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;177:3439–3447. doi: 10.4049/jimmunol.177.5.3439. [DOI] [PubMed] [Google Scholar]
- Lundstrom SL, Saluja R, Adner M, Haeggstrom JZ, Nilsson G, Wheelock CE. Lipid mediator metabolic profiling demonstrates differences in eicosanoid patterns in two phenotypically distinct mast cell populations. Journal of lipid research. 2013;54:116–126. doi: 10.1194/jlr.M030171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maceyka M, Spiegel S. Sphingolipid metabolites in inflammatory disease. Nature. 2014;510:58–67. doi: 10.1038/nature13475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Massberg S, Schaerli P, Knezevic-Maramica I, Kollnberger M, Tubo N, Moseman EA, Huff IV, Junt T, Wagers AJ, Mazo IB, von Andrian UH. Immunosurveillance by hematopoietic progenitor cells trafficking through blood, lymph, and peripheral tissues. Cell. 2007;131:994–1008. doi: 10.1016/j.cell.2007.09.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mechtcheriakova D, Wlachos A, Sobanov J, Kopp T, Reuschel R, Bornancin F, Cai R, Zemann B, Urtz N, Stingl G, Zlabinger G, Woisetschlager M, Baumruker T, Billich A. Sphingosine 1-phosphate phosphatase 2 is induced during inflammatory responses. Cellular signalling. 2007;19:748–760. doi: 10.1016/j.cellsig.2006.09.004. [DOI] [PubMed] [Google Scholar]
- 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;195:583–592. doi: 10.1084/jem.20020044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mellor EA, Frank N, Soler D, Hodge MR, Lora JM, Austen KF, Boyce JA. Expression of the type 2 receptor for cysteinyl leukotrienes (CysLT2R) by human mast cells: Functional distinction from CysLT1R. Proc Natl Acad Sci U S A. 2003;100:11589–11593. doi: 10.1073/pnas.2034927100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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;98:7964–7969. doi: 10.1073/pnas.141221498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mencia-Huerta JM, Lewis RA, Razin E, Austen KF. Antigen-initiated release of platelet-activating factor (PAF-acether) from mouse bone marrow-derived mast cells sensitized with monoclonal IgE. J Immunol. 1983;131:2958–2964. [PubMed] [Google Scholar]
- Metz M, Grimbaldeston MA, Nakae S, Piliponsky AM, Tsai M, Galli SJ. Mast cells in the promotion and limitation of chronic inflammation. Immunological reviews. 2007;217:304–328. doi: 10.1111/j.1600-065X.2007.00520.x. [DOI] [PubMed] [Google Scholar]
- Mitra P, Oskeritzian CA, Payne SG, Beaven MA, Milstien S, Spiegel S. Role of ABCC1 in export of sphingosine-1-phosphate from mast cells. Proc Natl Acad Sci U S A. 2006;103:16394–16399. doi: 10.1073/pnas.0603734103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Montuschi P, Peters-Golden ML. Leukotriene modifiers for asthma treatment. Clin Exp Allergy. 2010;40:1732–1741. doi: 10.1111/j.1365-2222.2010.03630.x. [DOI] [PubMed] [Google Scholar]
- Moon TC, Campos-Alberto E, Yoshimura T, Bredo G, Rieger AM, Puttagunta L, Barreda DR, Befus AD, Cameron L. Expression of DP2 (CRTh2), a Prostaglandin D2 Receptor, in Human Mast Cells. PloS one. 2014;9:e108595. doi: 10.1371/journal.pone.0108595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moriyama S, Takahashi N, Green JA, Hori S, Kubo M, Cyster JG, Okada T. Sphingosine-1-phosphate receptor 2 is critical for follicular helper T cell retention in germinal centers. The Journal of experimental medicine. 2014;211:1297–1305. doi: 10.1084/jem.20131666. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morrow JD, Guzzo C, Lazarus G, Oates JA, Roberts LJ., 2nd Improved diagnosis of mastocytosis by measurement of the major urinary metabolite of prostaglandin D2. The Journal of investigative dermatology. 1995;104:937–940. doi: 10.1111/1523-1747.ep12606209. [DOI] [PubMed] [Google Scholar]
- Murakami M, Matsumoto R, Austen KF, Arm JP. Prostaglandin endoperoxide synthase-1 and -2 couple to different transmembrane stimuli to generate prostaglandin D2 in mouse bone marrow-derived mast cells. The Journal of biological chemistry. 1994;269:22269–22275. [PubMed] [Google Scholar]
- Murakami M, Matsumoto R, Urade Y, Austen KF, Arm JP. c-kit ligand mediates increased expression of cytosolic phospholipase A2, prostaglandin endoperoxide synthase-1, and hematopoietic prostaglandin D2 synthase and increased IgE-dependent prostaglandin D2 generation in immature mouse mast cells. The Journal of biological chemistry. 1995;270:3239–3246. doi: 10.1074/jbc.270.7.3239. [DOI] [PubMed] [Google Scholar]
- Naclerio RM, Proud D, Togias AG, Adkinson NF, Jr., Meyers DA, Kagey-Sobotka A, Plaut M, Norman PS, Lichtenstein LM. Inflammatory mediators in late antigen-induced rhinitis. The New England journal of medicine. 1985;313:65–70. doi: 10.1056/NEJM198507113130201. [DOI] [PubMed] [Google Scholar]
- Nantel F, Fong C, Lamontagne S, Wright DH, Giaid A, Desrosiers M, Metters KM, O’Neill GP, Gervais FG. Expression of prostaglandin D synthase and the prostaglandin D2 receptors DP and CRTH2 in human nasal mucosa. Prostaglandins & other lipid mediators. 2004;73:87–101. doi: 10.1016/j.prostaglandins.2003.12.002. [DOI] [PubMed] [Google Scholar]
- Nilsson G, Metcalfe DD, Taub DD. Demonstration that platelet-activating factor is capable of activating mast cells and inducing a chemotactic response. Immunology. 2000;99:314–319. doi: 10.1046/j.1365-2567.2000.00972.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oliveira SH, Hogaboam CM, Berlin A, Lukacs NW. SCF-induced airway hyperreactivity is dependent on leukotriene production. American journal of physiology. Lung cellular and molecular physiology. 2001;280:L1242–1249. doi: 10.1152/ajplung.2001.280.6.L1242. [DOI] [PubMed] [Google Scholar]
- Olivera A, Urtz N, Mizugishi K, Yamashita Y, Gilfillan AM, Furumoto Y, Gu H, Proia RL, Baumruker T, Rivera J. IgE-dependent activation of sphingosine kinases 1 and 2 and secretion of sphingosine 1-phosphate requires Fyn kinase and contributes to mast cell responses. J Biol Chem. 2006;281:2515–2525. doi: 10.1074/jbc.M508931200. [DOI] [PubMed] [Google Scholar]
- Olivera A, Mizugishi K, Tikhonova A, Ciaccia L, Odom S, Proia RL, Rivera J. The sphingosine kinase-sphingosine-1-phosphate axis is a determinant of mast cell function and anaphylaxis. Immunity. 2007;26:287–297. doi: 10.1016/j.immuni.2007.02.008. [DOI] [PubMed] [Google Scholar]
- Olivera A. Unraveling the complexities of sphingosine-1-phosphate function: the mast cell model. Prostaglandins & other lipid mediators. 2008;86:1–11. doi: 10.1016/j.prostaglandins.2008.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Olivera A, Eisner C, Kitamura Y, Dillahunt S, Allende L, Tuymetova G, Watford W, Meylan F, Diesner SC, Li L, Schnermann J, Proia RL, Rivera J. Sphingosine kinase 1 and sphingosine-1-phosphate receptor 2 are vital to recovery from anaphylactic shock in mice. The Journal of clinical investigation. 2010;120:1429–1440. doi: 10.1172/JCI40659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Olivera A, Allende ML, Proia RL. Shaping the landscape: metabolic regulation of S1P gradients. Biochimica et biophysica acta. 2013a;1831:193–202. doi: 10.1016/j.bbalip.2012.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Olivera A, Dillahunt SE, Rivera J. Interrogation of sphingosine-1-phosphate receptor 2 function in vivo reveals a prominent role in the recovery from IgE and IgG-mediated anaphylaxis with minimal effect on its onset. Immunology letters. 2013b;150:89–96. doi: 10.1016/j.imlet.2013.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Olivera A, Kitamura Y, Wright LD, Allende ML, Chen W, Kaneko-Goto T, Yoshihara Y, Proia RL, Rivera J. Sphingosine-1-phosphate can promote mast cell hyper-reactivity through regulation of contactin-4 expression. Journal of leukocyte biology. 2013c;94:1013–1024. doi: 10.1189/jlb.0313163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Olivera A, Rivera J. Paradigm shifts in mast cell and basophil biology and function: an emerging view of immune regulation in health and disease. Methods in molecular biology. 2014;1192:3–31. doi: 10.1007/978-1-4939-1173-8_1. [DOI] [PubMed] [Google Scholar]
- Oskeritzian CA, Alvarez SE, Hait NC, Price MM, Milstien S, Spiegel S. Distinct roles of sphingosine kinases 1 and 2 in human mast-cell functions. Blood. 2008;111:4193–4200. doi: 10.1182/blood-2007-09-115451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oskeritzian CA, Price MM, Hait NC, Kapitonov D, Falanga YT, Morales JK, Ryan JJ, Milstien S, Spiegel S. Essential roles of sphingosine-1-phosphate receptor 2 in human mast cell activation, anaphylaxis, and pulmonary edema. The Journal of experimental medicine. 2010;207:465–474. doi: 10.1084/jem.20091513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Padawer J. Mast cells: extended lifespan and lack of granule turnover under normal in vivo conditions. Experimental and molecular pathology. 1974;20:269–280. doi: 10.1016/0014-4800(74)90059-8. [DOI] [PubMed] [Google Scholar]
- Pappu R, Schwab SR, Cornelissen I, Pereira JP, Regard JB, Xu Y, Camerer E, Zheng YW, Huang Y, Cyster JG, Coughlin SR. Promotion of lymphocyte egress into blood and lymph by distinct sources of sphingosine-1-phosphate. Science. 2007;316:295–298. doi: 10.1126/science.1139221. [DOI] [PubMed] [Google Scholar]
- Paruchuri S, Jiang Y, Feng C, Francis SA, Plutzky J, Boyce JA. Leukotriene E4 activates peroxisome proliferator-activated receptor gamma and induces prostaglandin D2 generation by human mast cells. J Biol Chem. 2008;283:16477–16487. doi: 10.1074/jbc.M705822200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Petersen LJ, Church MK, Skov PS. Platelet-activating factor induces histamine release from human skin mast cells in vivo, which is reduced by local nerve blockade. The Journal of allergy and clinical immunology. 1997;99:640–647. doi: 10.1016/s0091-6749(97)70026-5. [DOI] [PubMed] [Google Scholar]
- Pettipher R. The roles of the prostaglandin D(2) receptors DP(1) and CRTH2 in promoting allergic responses. British journal of pharmacology. 2008;153(Suppl 1):S191–199. doi: 10.1038/sj.bjp.0707488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pettipher R, Whittaker M. Update on the development of antagonists of chemoattractant receptor-homologous molecule expressed on Th2 cells (CRTH2). From lead optimization to clinical proof-of-concept in asthma and allergic rhinitis. Journal of medicinal chemistry. 2012;55:2915–2931. doi: 10.1021/jm2013997. [DOI] [PubMed] [Google Scholar]
- Prescott SM, Zimmerman GA, Stafforini DM, McIntyre TM. Platelet-activating factor and related lipid mediators. Annual review of biochemistry. 2000;69:419–445. doi: 10.1146/annurev.biochem.69.1.419. [DOI] [PubMed] [Google Scholar]
- Price MM, Kapitonov D, Allegood J, Milstien S, Oskeritzian CA, Spiegel S. Sphingosine-1-phosphate induces development of functionally mature chymase-expressing human mast cells from hematopoietic progenitors. FASEB J. 2009;23:3506–3515. doi: 10.1096/fj.08-128900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Prieschl EE, Csonga R, Novotny V, Kikuchi GE, Baumruker T. The balance between sphingosine and sphingosine-1-phosphate is decisive for mast cell activation after FceRI triggering. J. Exp. Med. 1999;190:1–8. doi: 10.1084/jem.190.1.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rajakariar R, Hilliard M, Lawrence T, Trivedi S, Colville-Nash P, Bellingan G, Fitzgerald D, Yaqoob MM, Gilroy DW. Hematopoietic prostaglandin D2 synthase controls the onset and resolution of acute inflammation through PGD2 and 15-deoxyDelta12 14 PGJ2. Proceedings of the National Academy of Sciences of the United States of America. 2007;104:20979–20984. doi: 10.1073/pnas.0707394104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reines I, Kietzmann M, Mischke R, Tschernig T, Luth A, Kleuser B, Baumer W. Topical application of sphingosine-1-phosphate and FTY720 attenuate allergic contact dermatitis reaction through inhibition of dendritic cell migration. The Journal of investigative dermatology. 2009;129:1954–1962. doi: 10.1038/jid.2008.454. [DOI] [PubMed] [Google Scholar]
- Rivera J, Gilfillan AM. Molecular regulation of mast cell activation. J. Allergy Clin. Immunol. 2006;117:1214–1225. doi: 10.1016/j.jaci.2006.04.015. [DOI] [PubMed] [Google Scholar]
- Rivera J, Proia RL, Olivera A. The alliance of sphingosine-1-phosphate and its receptors in immunity. Nature reviews. Immunology. 2008;8:753–763. doi: 10.1038/nri2400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roviezzo F, Brancaleone V, De Gruttola L, Vellecco V, Bucci M, D’Agostino B, Cooper D, Sorrentino R, Perretti M, Cirino G. Sphingosine-1-phosphate modulates vascular permeability and cell recruitment in acute inflammation in vivo. The Journal of pharmacology and experimental therapeutics. 2011;337:830–837. doi: 10.1124/jpet.111.179168. [DOI] [PubMed] [Google Scholar]
- Ryan JJ, Spiegel S. The role of sphingosine-1-phosphate and its receptors in asthma. Drug news & perspectives. 2008;21:89–96. doi: 10.1358/dnp.2008.21.2.1188195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sanchez T, Hla T. Structural and functional characteristics of S1P receptors. J Cell Biochem. 2004;92:913–922. doi: 10.1002/jcb.20127. [DOI] [PubMed] [Google Scholar]
- Sanchez T, Skoura A, Wu MT, Casserly B, Harrington EO, Hla T. Induction of vascular permeability by the sphingosine-1-phosphate receptor-2 (S1P2R) and its downstream effectors ROCK and PTEN. Arteriosclerosis, thrombosis, and vascular biology. 2007;27:1312–1318. doi: 10.1161/ATVBAHA.107.143735. [DOI] [PubMed] [Google Scholar]
- Schaper K, Dickhaut J, Japtok L, Kietzmann M, Mischke R, Kleuser B, Baumer W. Sphingosine-1-phosphate exhibits anti-proliferative and anti-inflammatory effects in mouse models of psoriasis. Journal of dermatological science. 2013;71:29–36. doi: 10.1016/j.jdermsci.2013.03.006. [DOI] [PubMed] [Google Scholar]
- Scher JU, Pillinger MH. The anti-inflammatory effects of prostaglandins. Journal of investigative medicine: the official publication of the American Federation for Clinical Research. 2009;57:703–708. doi: 10.2310/JIM.0b013e31819aaa76. [DOI] [PubMed] [Google Scholar]
- Schulze T, Golfier S, Tabeling C, Rabel K, Graler MH, Witzenrath M, Lipp M. Sphingosine-1-phospate receptor 4 (S1P(4)) deficiency profoundly affects dendritic cell function and TH17-cell differentiation in a murine model. FASEB journal: official publication of the Federation of American Societies for Experimental Biology. 2011;25:4024–4036. doi: 10.1096/fj.10-179028. [DOI] [PubMed] [Google Scholar]
- Schwab SR, Pereira JP, Matloubian M, Xu Y, Huang Y, Cyster JG. Lymphocyte sequestration through S1P lyase inhibition and disruption of S1P gradients. Science. 2005;309:1735–1739. doi: 10.1126/science.1113640. [DOI] [PubMed] [Google Scholar]
- Seo EY, Park GT, Lee KM, Kim JA, Lee JH, Yang JM. Identification of the target genes of atopic dermatitis by real-time PCR. The Journal of investigative dermatology. 2006;126:1187–1189. doi: 10.1038/sj.jid.5700234. [DOI] [PubMed] [Google Scholar]
- Serhan CN, Chiang N, Van Dyke TE. Resolving inflammation: dual anti-inflammatory and pro-resolution lipid mediators. Nature reviews. Immunology. 2008;8:349–361. doi: 10.1038/nri2294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Serra M, Saba JD. Sphingosine 1-phosphate lyase, a key regulator of sphingosine 1-phosphate signaling and function. Advances in enzyme regulation. 2010;50:349–362. doi: 10.1016/j.advenzreg.2009.10.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Serra-Pages M, Olivera A, Torres R, Picado C, de Mora F, Rivera J. E-prostanoid 2 receptors dampen mast cell degranulation via cAMP/PKA-mediated suppression of IgE-dependent signaling. Journal of leukocyte biology. 2012;92:1155–1165. doi: 10.1189/jlb.0212109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spiegel S, Milstien S. The outs and the ins of sphingosine-1-phosphate in immunity. Nature reviews. Immunology. 2011;11:403–415. doi: 10.1038/nri2974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Strait RT, Morris SC, Yang M, Qu XW, Finkelman FD. Pathways of anaphylaxis in the mouse. The Journal of allergy and clinical immunology. 2002;109:658–668. doi: 10.1067/mai.2002.123302. [DOI] [PubMed] [Google Scholar]
- Tager AM, Luster AD. BLT1 and BLT2: the leukotriene B(4) receptors. Prostaglandins, leukotrienes, and essential fatty acids. 2003;69:123–134. doi: 10.1016/s0952-3278(03)00073-5. [DOI] [PubMed] [Google Scholar]
- Taha TA, Hannun YA, Obeid LM. Sphingosine kinase: biochemical and cellular regulation and role in disease. J Biochem Mol Biol. 2006;39:113–131. doi: 10.5483/bmbrep.2006.39.2.113. [DOI] [PubMed] [Google Scholar]
- Taketomi Y, Ueno N, Kojima T, Sato H, Murase R, Yamamoto K, Tanaka S, Sakanaka M, Nakamura M, Nishito Y, Kawana M, Kambe N, Ikeda K, Taguchi R, Nakamizo S, Kabashima K, Gelb MH, Arita M, Yokomizo T, Nakamura M, Watanabe K, Hirai H, Nakamura M, Okayama Y, Ra C, Aritake K, Urade Y, Morimoto K, Sugimoto Y, Shimizu T, Narumiya S, Hara S, Murakami M. Mast cell maturation is driven via a group III phospholipase A2-prostaglandin D2-DP1 receptor paracrine axis. Nature immunology. 2013;14:554–563. doi: 10.1038/ni.2586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thomas D, Suo J, Ulshofer T, Jordan H, de Bruin N, Scholich K, Geisslinger G, Ferreiros N. Nano-LC-MS/MS for the quantitation of prostanoids in immune cells. Analytical and bioanalytical chemistry. 2014;406:7103–7116. doi: 10.1007/s00216-014-8134-8. [DOI] [PubMed] [Google Scholar]
- Torres R, Picado C, de Mora F. The PGE2-EP2-mast cell axis: An antiasthma mechanism. Molecular immunology. 2014 doi: 10.1016/j.molimm.2014.03.007. [DOI] [PubMed] [Google Scholar]
- Trivedi SG, Newson J, Rajakariar R, Jacques TS, Hannon R, Kanaoka Y, Eguchi N, Colville-Nash P, Gilroy DW. Essential role for hematopoietic prostaglandin D2 synthase in the control of delayed type hypersensitivity. Proceedings of the National Academy of Sciences of the United States of America. 2006;103:5179–5184. doi: 10.1073/pnas.0507175103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tsoupras AB, Iatrou C, Frangia C, Demopoulos CA. The implication of platelet activating factor in cancer growth and metastasis: potent beneficial role of PAF-inhibitors and antioxidants. Infectious disorders drug targets. 2009;9:390–399. doi: 10.2174/187152609788922555. [DOI] [PubMed] [Google Scholar]
- Tsujimura Y, Obata K, Mukai K, Shindou H, Yoshida M, Nishikado H, Kawano Y, Minegishi Y, Shimizu T, Karasuyama H. Basophils play a pivotal role in immunoglobulin-G-mediated but not immunoglobulin-E-mediated systemic anaphylaxis. Immunity. 2008;28:581–589. doi: 10.1016/j.immuni.2008.02.008. [DOI] [PubMed] [Google Scholar]
- Urtz N, Olivera A, Bofill-Cardona E, Csonga R, Billich A, Mechtcheriakova D, Bornancin F, Woisetschlager M, Rivera J, Baumruker T. Early activation of sphingosine kinase in mast cells and recruitment to FcepsilonRI are mediated by its interaction with Lyn kinase. Mol Cell Biol. 2004;24:8765–8777. doi: 10.1128/MCB.24.19.8765-8777.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vadas P, Gold M, Perelman B, Liss GM, Lack G, Blyth T, Simons FE, Simons KJ, Cass D, Yeung J. Platelet-activating factor, PAF acetylhydrolase, and severe anaphylaxis. The New England journal of medicine. 2008;358:28–35. doi: 10.1056/NEJMoa070030. [DOI] [PubMed] [Google Scholar]
- Weller CL, Collington SJ, Brown JK, Miller HR, Al-Kashi A, Clark P, Jose PJ, Hartnell A, Williams TJ. Leukotriene B4, an activation product of mast cells, is a chemoattractant for their progenitors. J Exp Med. 2005;201:1961–1971. doi: 10.1084/jem.20042407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xue L, Gyles SL, Wettey FR, Gazi L, Townsend E, Hunter MG, Pettipher R. Prostaglandin D2 causes preferential induction of proinflammatory Th2 cytokine production through an action on chemoattractant receptor-like molecule expressed on Th2 cells. Journal of immunology. 2005;175:6531–6536. doi: 10.4049/jimmunol.175.10.6531. [DOI] [PubMed] [Google Scholar]
- Yoshimura-Uchiyama C, Iikura M, Yamaguchi M, Nagase H, Ishii A, Matsushima K, Yamamoto K, Shichijo M, Bacon KB, Hirai K. Differential modulation of human basophil functions through prostaglandin D2 receptors DP and chemoattractant receptor-homologous molecule expressed on Th2 cells/DP2. Clinical and experimental allergy: journal of the British Society for Allergy and Clinical Immunology. 2004;34:1283–1290. doi: 10.1111/j.1365-2222.2004.02027.x. [DOI] [PubMed] [Google Scholar]
- Zhang G, Yang L, Kim GS, Ryan K, Lu S, O’Donnell RK, Spokes K, Shapiro N, Aird WC, Kluk MJ, Yano K, Sanchez T. Critical role of sphingosine-1-phosphate receptor 2 (S1PR2) in acute vascular inflammation. Blood. 2013;122:443–455. doi: 10.1182/blood-2012-11-467191. [DOI] [PMC free article] [PubMed] [Google Scholar]


