Basophilic granulocytes (usually referred to as basophils) are a small population of peripheral blood leukocytes containing cytoplasmatic granules that stain with basophilic dyes. They were first described in 1879 by Paul Ehrlich,1 who 1 year earlier had found a morphologically similar cell type present in tissues that he termed Mastzellen.2 Based on their similarity to mast cells, basophils have often been considered (and neglected) as minor and possibly redundant “circulating mast cells.” Like mast cells, basophils possess high-affinity immunoglobulin (Ig) E receptors (FcεRI) that are cross-linked upon engagement of receptor-bound IgE with corresponding antigens (“allergens”), resulting in the release of a number of mediators that are in part common for both cell types. Until recently, it has been very difficult for most laboratories to obtain basophils without major contaminating cell populations, because the percentage of basophils in peripheral blood is low (< 1%) and they share physicochemical properties with other blood cells. This lack of satisfactory purification protocols has considerably hampered basophil research and negatively affected the interest in this cell type. Nevertheless, recent findings have provided new insights into the possible role of basophils in allergic disease and immunity to pathogens. Most notably, the discovery that basophils rapidly produce large amounts of the regulatory cytokines interleukin (IL)-4 3,4 and IL-13,5-7 together with the constitutive expression of CD40L 8,9 and CCR310 on their surface, has fueled speculations that extend beyond their recognized role as effector cells in IgE-mediated reactions. As discussed in this review, the increasing knowledge about the chemokine network has provided a robust framework for understanding the mechanisms underlying basophil recruitment to the tissues in allergic diseases. This development has been instrumental in identifying very promising new targets for the treatment of asthma and other allergic diseases. We also review recent aspects regarding basophil growth and development and their role in allergic late-phase reactions (LPRs). Furthermore, we have included a section on basophil signal transduction, which expands our understanding of the mechanisms operating downstream of the high-affinity IgE receptor and are also expected to yield new and better targets for pharmacologic intervention. Finally, we discuss the possible roles of basophils in innate immunity and finish with a section on the currently available tools for basophil research.

Although the phenotype of mature basophils has been extensively studied,11,12 the early stages of basophil maturation and their relationship to other cell lineages are not well understood. Basophils, as well as mast cells, monocytes, eosinophils, and neutrophils, are thought to arise from CD34+ progenitors found in cord blood, peripheral blood, and the bone marrow. Basophils, in particular, have been suggested to evolve from CD34+/IL-3Rα+/IL-5+eosinophil/basophil progenitors,13 as supported by the occurrence of granulocytes with a hybrid eosinophil/basophil phenotype in patients with chronic or acute myelogenous leukemia or in cell culture.14-16 

Although mast cell progenitors have been documented as a separate lineage, eg, characterized as CD34+/CD38+17 or CD34+/c-kit+/CD13+,18most recently a new monoclonal antibody (97A6) has been described as specific for mature mast cells and basophils and their progenitors.19 Monoclonal antibody 97A6 did not react with any other hematopoietic or nonhematopoietic cell type. The epitope recognized by 97A6 may therefore be associated with a commitment of the CD34+ precursor to a mast cell or basophil lineage distinct from other lineages. The possibility of a common mast cell or basophil lineage also arises from the surprising observation that basophils with phenotypic features characteristic of mast cells (presence of tryptase, chymase, c-kit, carboxypeptidase A) can be found in patients with asthma, allergy, or allergic-drug reactions.20Therefore, the currently predominant concept that mast cells and basophils originate from separate lineages17,21,22 may have to be revised.

Various protocols using different combinations of cytokines and growth factors have been described for the in vitro cultivation of basophils from blood or bone marrow progenitors,23-25 with the presence of exogenous IL-3 being a common prerequisite for the successful development and maturation of basophils. Other growth factors for basophils are IL-5,26 granulocyte-macrophage colony-stimulating factor (GM-CSF),27 transforming growth factor-β (TGF-β), and nerve growth factor (NGF).28There is a general consensus, however, that the main growth and differentiation factor for basophils is IL-3,24,29 whereas the growth and development of mast cells requires the presence of stem cell factor (SCF).30 In conformity with their relative cytokine requirements, mature basophils can be distinguished phenotypically from mast cells by their differential surface expression of IL-3 α chain (CD123, found on basophils and other cells but not mast cells) and SCF receptor (c-kit/CD117, strongly expressed on mast cells but not, or only weakly, on mature basophils).

It is well established that mast cells, which are found in the tissues in the proximity of small blood vessels and postcapillary venules, play a key role in the early phase of IgE-mediated allergic reactions.31 Unlike mast cells, which differentiate and mature in the tissues, mature basophils are found in the circulatory system, where they constitute less than 1% of circulating leukocytes. Basophils, eosinophils, and Th2 lymphocytes are recruited to the site of inflammation during LPRs. Because until recently specific markers for immunohistochemical detection of basophils were not available (see “Tools for basophil research”), the participation of basophils during LPRs has been documented mainly by indirect means,32-34 ie, by determining the pattern of mast cell– or basophil-specific mediators like histamine (derived from both), prostaglandin D2 (from mast cells), or leukotriene C4 (LTC4; from basophils). It is now well established that basophils are rapidly recruited to the skin,35 lung,36 or nose37 after allergen challenge.

More recently, a better understanding of the processes leading to inflammatory cell recruitment in LPRs has sprung up from the study of chemokines and their receptor counterparts, identifying potentially important new targets for the treatment of asthma and other allergic diseases.38-40 These studies complement previous knowledge regarding the molecules involved in basophil adhesion to the endothelium,41-43 a critical step in the process of extravasation. The aim of the following section is to highlight the potential roles of basophils in LPRs. We use the new nomenclature for chemokines proposed by the Keystone Chemokine Conference (Keystone, CO, January 18-23, 1999).40 

Several reports have pinpointed eotaxin (CCL11) and the eotaxin receptor (CCR3) as key molecules in the selective recruitment of eosinophils to the lung during allergic airway inflammation,44 both in animal models45-49 and asthmatic patients.50-53 Sensitized mice with a disrupted eotaxin gene showed a 70% reduction in the number of eosinophils recruited by antigen in comparison with the identically treated wild-type mice.54 Enhanced eotaxin expression in allergic inflammation may also account for the influx of basophils, which strongly express CCR3 on their surface and respond to eotaxin in vitro.10,55 In addition, human basophils also respond to other chemokines such as the CC chemokines eotaxin-2 (CCL24), RANTES, MCP-3 (CCL7), MCP-1, and MIP-1α,56,57 which also play a role in antigen-induced tissue recruitment of eosinophils.58-60 Moreover, basophils may be responsive to eotaxin-3 (CCL26),61,62 the most recent addition to a growing list of CCR3 agonists. Although it remains to be proven, it is likely that the presence of CCR3 and other CC chemokine receptors shared between eosinophils and basophils account for the previously demonstrated strong temporal association of their influx into tissues such as the nose37,63 or the lung36 during LPRs.

A further insight regarding tissue influx of basophils and their subsequent activation is also given by the actions of hematopoietic cytokines such as IL-3, IL-5, and GM-CSF. An increased expression of these Th2 lymphocyte–derived cytokines in allergen-induced cutaneous LPRs in atopics was clearly demonstrated by Kay and colleagues,64 and these cytokines have also been shown to facilitate basophil migration.65-67 In addition, IL-3, IL-5, and GM-CSF also enhance mediator release following either IgE-dependent stimulation or activation due to platelet-activating factor, C5a, and C3a. Similar properties have additionally been described for the neurotrophic cytokine NGF.68 Moreover, increased levels of NGF have recently been reported in allergic rhinitis69 and asthma.70 

IL-3, IL-5, GM-CSF, and NGF have similar efficacies with respect to potentiating IgE-mediated basophil histamine and LTC4releases, and they bind to receptors of the same subfamily but, of these, IL-3 is the most potent71 and its effects on basophils have been appreciated for some time.72,73 In vitro, IL-3 alone causes little mediator release by itself, with the exception of IL-13 production in some donors. However, a short preincubation or coculture of basophils with IL-3 causes significant enhancement of histamine and LTC4 release to a number of immunologic and nonimmunologic stimuli such as anti-IgE, C5a, C3a, the eosinophil product major basic protein, and platelet-activating factor.74-77 Moreover, studies by Ochensberger78 demonstrate that LTC4 synthesis in basophils primed by IL-3 before stimulation with C5a is continuously maintained over many hours. This clearly highlights the ability of basophils to maintain a state of activity over longer periods than previously anticipated. Additionally, these authors showed an elevated production of IL-4 and IL-13 from basophils under the same conditions,78 which may exacerbate LPRs because of the up-regulatory effects of these cytokines on vascular cell adhesion molecule-1 (VCAM-1), thus promoting further influx of inflammatory cells (discussed in detail below).

The ability of basophils to continuously produce LTC4 and cytokines may also be particularly important in maintaining chronic allergic inflammatory diseases, such as asthma. Furthermore, because IL-3 can, in conjunction with C5a, cause basophil mediator release independently of IgE-receptor cross-linking, this suggests a role for basophils in nonallergic inflammatory diseases caused by either bacterial infections or IgG immune complexes. In addition to the combined effects of C5a and IL-3, C5a by itself is a highly potent and efficacious inducer of histamine release from basophils79following binding to the C5a receptor, CD88.80 In contrast, most human mast cell subtypes do not express CD88 and are unresponsive to C5a except skin mast cells.80 Moreover, unlike basophils, mature human mast cells, including those of the skin, also do not express receptors for IL-3 and GM-CSF.81 This clearly suggests that, compared with mast cells, basophils are more sensitive to the effects of immunomodulatory cytokines and proinflammatory factors, which are likely to be in abundance in both LPRs and other (nonallergic) inflammatory conditions.

Unfortunately, the interplay of these various basophil activators, in terms of the net clinical outcome of their combined effects, in allergic disease remains to be elucidated. However, the importance of IL-3 in vivo as both a modulator of basophil growth and activation as well as its ability to cause clinical symptoms of allergic inflammation was shown by van Gils et al.82 Using rhesus monkeys treated with the IL-3, the authors observed a clear increase in both basophil numbers together with an up-regulation of IL-3 receptors without a concomitant rise of IL-3R+ monocytes or mast cells.82 Moreover, the same authors also demonstrated that IL-3 treatment caused severe hypersensitivity reactions including urticaria, vomiting, diarrhea, edema, arthritis, and stimulation of hemopoiesis.83 Given the above evidence, IL-3 and related hematopoietic growth factors, as well as NGF, potentially play a pivotal role in exacerbating LPRs in addition to CC chemokines describedearlier.

Another indirect line of evidence that reveals basophils as potential major regulatory cells in allergic inflammation comes from the study of their cytokine profile. As described earlier, human basophils are an important source of the proallergic cytokines IL-4 and IL-13. Kasaian et al84 have shown that allergen-induced IL-4 from primary cultures of unfractionated peripheral blood leukocytes of atopic patients is mainly basophil-derived. This finding has been confirmed and extended to IL-13 by Devouassoux and coworkers,85 who found that basophils are the main source of both cytokines early after allergen stimulation of peripheral blood cells. IL-4 is of central importance for the recruitment of inflammatory cells to the tissues, because it induces the expression of the adhesion molecule VCAM-1 on endothelial cells in vitro42 and it recruits eosinophils in vivo, eg, when injected in rodents.86,87 Transgenic mice expressing IL-4 in the lung under the control of a lung-specific promoter showed eosinophil infiltrations in the airways.88Recently, Hickey et al89 have used intravital microscopy to demonstrate that the VLA (very late antigen)-4/VCAM-1 interaction is necessary and sufficient for IL-4–induced eosinophil recruitment in mice. On the other hand, it has been shown that IL-4, especially in combination with tumor necrosis factor (TNF)–α, induces the production and release of eotaxin by human dermal fibroblasts;90 moreover, eotaxin-3, but not eotaxin, is up-regulated in vascular endothelial cells by IL-4.62 The induction of eotaxins by IL-4 could represent a strong positive feedback loop operating in eosinophilic skin diseases (eg, atopic dermatitis), with the induced eotaxins recruiting more eosinophils, basophils, and Th2 lymphocytes and the subsequent release of more IL-4 upon activation (Figure 1). Interestingly, eotaxin can potentiate the antigen-dependent IL-4 production by basophils91 without having a direct effect on basophils by itself,78 thus generating a further potential amplification loop in allergic inflammation. The aforementioned proallergic properties of IL-4 therefore make the identification of its cellular source(s) an important prerequisite for the successful therapy of asthma and other allergic diseases.

Fig. 1.

LPR scenario emphasizing the putative regulatory roles of basophils in allergic inflammation.

Eotaxin (CCL11) (A) derived from epithelial cells,184eosinophils,51 or mast cells185 can be detected in the lungs of asthmatic patients50-53 and recruits cells expressing the eotaxin receptor (CCR3), ie, eosinophils,186,187 basophils,10,55 Th2 cells,188,189 and mast cells.190,191Recruited basophils are a major source of the regulatory cytokines IL-43,4 and IL-13.5-7 There are several potential amplification loops, such as the up-regulation of eotaxin in airway epithelial cells97 (B) and VCAM-1 on vascular endothelium (C) by IL-1396 or IL-442 or, also, the up-regulation of eotaxin by IL-4 in fibroblasts.90Eotaxin has also been shown to potentiate the IL-4 production by activated basophils91 (D). Furthermore, basophils can deliver the signals necessary for switching B cells to IgE (E), ie, IL-4, IL-13, and CD40L.8,9 The increased IgE production is thought to up-regulate the expression of FcεRI on basophils and mast cells103,104 (F).

Fig. 1.

LPR scenario emphasizing the putative regulatory roles of basophils in allergic inflammation.

Eotaxin (CCL11) (A) derived from epithelial cells,184eosinophils,51 or mast cells185 can be detected in the lungs of asthmatic patients50-53 and recruits cells expressing the eotaxin receptor (CCR3), ie, eosinophils,186,187 basophils,10,55 Th2 cells,188,189 and mast cells.190,191Recruited basophils are a major source of the regulatory cytokines IL-43,4 and IL-13.5-7 There are several potential amplification loops, such as the up-regulation of eotaxin in airway epithelial cells97 (B) and VCAM-1 on vascular endothelium (C) by IL-1396 or IL-442 or, also, the up-regulation of eotaxin by IL-4 in fibroblasts.90Eotaxin has also been shown to potentiate the IL-4 production by activated basophils91 (D). Furthermore, basophils can deliver the signals necessary for switching B cells to IgE (E), ie, IL-4, IL-13, and CD40L.8,9 The increased IgE production is thought to up-regulate the expression of FcεRI on basophils and mast cells103,104 (F).

Close modal

Recent evidence has implicated the pleiotropic cytokine IL-13, which is also produced in large amounts by activated basophils, as a key mediator of allergic asthma. In humans, IL-13 is secreted following local allergen challenge in subjects with mild atopic asthma.92 Work performed by 2 independent laboratories, using an IL-13R–Fc fusion protein that specifically neutralizes IL-13, has indicated that this cytokine is necessary and sufficient for establishment of an asthmatic phenotype in a murine model.93,94 Administration of soluble recombinant IL-13 was effective in inducing airway hyperresponsiveness and in generating a significant recruitment of eosinophils to the lung, as shown by their appearance in bronchoalveolar lavages.93 In vitro, IL-13 induces recruitment and prolongs the survival of eosinophils,95 up-regulates VCAM-1 expression on vascular endothelium,96 and is a very potent inducer of eotaxin in airway epithelial cells.97 The importance of IL-13 in the pathogenesis of asthma is also corroborated by a recent study using transgenic mice that express IL-13 selectively in the lung.98 The authors of this study found that transgenic mice, in the absence of a specific sensitization, developed a phenotype very similar to the asthmatic phenotype, including eosinophil infiltration, up-regulation of eotaxin, mucus hypersecretion by goblet cells, subepithelial airway fibrosis, and airway hyperresponsiveness to methacholine.98 

Finally, basophils have also been proposed to play a key role in allergy by directly inducing the switch to the IgE isotype in B cells independently of T cells (Figure 1). Gauchat et al have shown that the basophil cell line KU812, as well as purified peripheral blood basophils, the mast cell line HMC-1, and human lung mast cells, can provide the necessary signals for IgE production in vitro, ie, IL-4, IL-13, and CD40L.9 Basophils generated from human umbilical cord blood mononuclear cells, after cultivation in the presence of appropriate cytokines, expressed detectable levels of CD40L and induced IgG4 and IgE synthesis in B cells when stimulated with allergen.8 The induction of IgE synthesis was completely abrogated by neutralizing IL-4 and IL-13 with monoclonal antibodies or CD40L with soluble CD40.8 

Taken together, the data about the involvement of IL-4, IL-13, and eotaxin as key players in the pathogenesis of asthma, together with the proven infiltration and activation of basophils in the tissues during LPRs, makes it reasonable to assume that basophils, as a major source of IL-4 and IL-13 and by constitutively expressing CCR3 and CD40L on their surface, may play an important but underestimated role in the pathology of asthma and other allergic conditions. The knowledge about the relative roles and contribution of basophils, eosinophils, mast cells, and T cells to these processes as well as about their interplay32 is still preliminary and needs further refinement.

The effector role of basophils in IgE-mediated allergy is well known to depend on cross-linking of FcεRI-bound IgE by allergens. The diversity of basophil mediators that are released following IgE-receptor cross-linking is accomplished by a highly complex and interrelated network of secondary messengers. Their specific roles in controlling mediator release are not nearly as well characterized for human basophils compared with their counterparts from other species or cell lines. Several examples given below show that a mere extrapolation of observations derived from basophil lines or basophils from other species does not clearly reflect the situation in primary human basophils. Although it may be obvious, this point must be stressed because future successful pharmacologic approaches to reducing basophil mediator production hinge crucially on understanding basophil biology taken from studies on primary basophils.

Early IgE-dependent signaling events in basophils

High-affinity IgE-receptor (FcεRI) cross-linking culminates in 3 main results. The first, and most rapid, is fusion of basophil granules to the plasma membrane resulting in histamine release99; the second is an activation of lipid metabolism and subsequent LTC4 production; and the third is synthesis of immunomodulatory cytokines. The initial signaling events following FcεRI cross-linking are currently modeled on studies largely carried out on rodent mast cell lines (such as RBL-2H3 cells), where phosphorylation of tyrosine, serine, and threonine residues contained in the β and γ chains of FcεRI receptor have been observed (Figure 2). In human basophils, as in RBL-2H3 cells, this has been shown to involve Lyn and subsequent recruitment of SH-2 domain–containing proteins such as Syk and other ZAP-70–related protein tyrosine kinases.101 The possibility that Syk may be involved in IgE-dependent basophil signaling was recently also demonstrated by Kepley and coworkers,102 who showed that nonreleaser human basophils, occurring in about 20% of the population, have a deficiency of this protein. In mast cells, most of the known major regulatory proteins known to be involved in FcεRI activation are downstream of Syk and thus crucially hinged to Syk activity. These include PLCγ1 and PLCγ2, Shc, Vav, mitogen-activated protein (MAP) kinases, phosphatidylinositol (PI)-3 kinase, and JNK, among others, although this has yet to be conclusively demonstrated for human basophils.

Fig. 2.

Schematic diagram of possible IgE-mediated signaling pathways in basophils.

Following high-affinity IgE-receptor cross-linking, various tyrosine kinases are activated, including Lyn and Syk, which consequently lead to the activation of PI 3-kinases, ERK-associated MAP kinases (Vav, Ras, Raf1, MEK), and PLC. PI 3-kinase targets include both p38 MAP kinase (p38) and Rac/Rho GTPases (Rac/Rho), which are involved in cytokine production. Further, Rac/Rho affect the cytoskeletal processes100 during degranulation as well as extracellular signal–related kinase-activating kinase (MEK). IgE-dependent ERK activation in basophils is largely limited to controlling LTC4 generation; PKC, activated by diacylglycerol (DAG) as a result of hydrolysis of PIP2 by PLC, is involved in degranulation. PKC may also affect cytokine transcription, although the mechanism is not yet clear. The release of calcium from intracellular stores by IP3 together with the influx of the ion through calcium channels affects degranulation, PLA2 translocation, and calcineurin activity. Thicker print is used to highlight what is currently known in human basophils. The remaining has been extrapolated from studies with rodent mast cells or cell lines.

Fig. 2.

Schematic diagram of possible IgE-mediated signaling pathways in basophils.

Following high-affinity IgE-receptor cross-linking, various tyrosine kinases are activated, including Lyn and Syk, which consequently lead to the activation of PI 3-kinases, ERK-associated MAP kinases (Vav, Ras, Raf1, MEK), and PLC. PI 3-kinase targets include both p38 MAP kinase (p38) and Rac/Rho GTPases (Rac/Rho), which are involved in cytokine production. Further, Rac/Rho affect the cytoskeletal processes100 during degranulation as well as extracellular signal–related kinase-activating kinase (MEK). IgE-dependent ERK activation in basophils is largely limited to controlling LTC4 generation; PKC, activated by diacylglycerol (DAG) as a result of hydrolysis of PIP2 by PLC, is involved in degranulation. PKC may also affect cytokine transcription, although the mechanism is not yet clear. The release of calcium from intracellular stores by IP3 together with the influx of the ion through calcium channels affects degranulation, PLA2 translocation, and calcineurin activity. Thicker print is used to highlight what is currently known in human basophils. The remaining has been extrapolated from studies with rodent mast cells or cell lines.

Close modal

Although several of the early IgE-dependent signaling events in basophils are known, there are currently no pharmacologic approaches to the treatment of allergy that specifically modulate very early signaling events following cross-linking with allergen. However, the molecules known to be involved in early post–cross-linking are also used by other immunoreceptor systems affecting many other immune cell functions, thus making selective pharmacologic intervention difficult. Therefore, an alternative strategy is to inhibit either FcεRI expression itself or the binding of IgE molecules to the receptor. It has long been observed that there is a positive correlation between circulating IgE antibodies and FcεRI expression in basophils,103,104 and recent evidence clearly demonstrates that IgE does directly induce high-affinity IgE-receptor expression by acting through FcεRI itself.104 Thus, basophil (and mast cell) function may significantly be down-regulated by disrupting the binding of IgE to its receptor at the sensitization stage. This has been successfully achieved in atopic patients treated with a monoclonal anti-IgE antibody (rhuMAb-E25), which prevents IgE binding to the FcεRI α chain without cross-linking high-affinity receptor-bound IgE,105 and is a promising new therapeutic advancement in the treatment of allergic disorders.

PI 3-kinase is a universal regulator of basophil mediator release

PI 3-kinase is composed of a p85 regulatory subunit and a 110-kd catalytic domain that catalyses the reaction of membrane PdtIns4,5P2 principally to PdtIns3,4,5P3. Indirect observations using specific PI 3-kinase antagonists suggest that PI 3-kinase plays a crucial role in the release of all major basophil mediator types, suggesting that this enzyme is closely associated with initial events in IgE-receptor signaling.106,107 We have recently shown that the PI 3-kinase inhibitors wortmannin and LY-294003 almost completely abrogate the release of histamine, LTC4, IL-4, and IL-13 in human basophils stimulated with anti-IgE.107Moreover, inhibition of both IL-4 and IL-13 production did not result in increased cellular contents of the cytokines, showing that PI 3-kinase blockade is not restricted to impairing secretory events.

With respect to mast cell and basophil degranulation, there is a universal consensus that PI 3-kinase inhibitors abolish granule-associated release.108-112 However, in regard to LTC4 production, Marquardt et al reported that the PI 3-kinase inhibitor wortmannin had no effect on the production of the eicosanoid in murine mast cells.109 The same authors also failed to detect any effects of this compound on cytokine release, whereas other reports have described inhibition of either leukotriene synthesis or TNF-α production.113,114 These differences show that a marked heterogeneity exists between basophils and various mast cell lineages to PI 3-kinase signaling.

Extracellular signal–related kinases control basophil LTC4 production but not histamine or cytokine release

The extracellular signal–related kinases (ERKs) consist of 2 forms, ERK-1 and ERK-2 (sometimes referred to as p44 and p42 MAP kinases), which are considerably homologous to another in both structure and function.115 ERKs are activated after FcεRI cross-linking via Vav, Ras-Raf1, and MEK1 and activate phospholipase A2 (PLA2) as well as several transcription factors (Figure 2). The role of ERKs in basophils, however, has only recently been partially investigated. Studies from our own laboratory and from Miura et al116 have shown that ERK phosphorylation occurs within 3 to 10 minutes of basophil stimulation, after which there is a decrease to basal levels. The MEK inhibitor PD-098059 abolishes both anti-IgE–induced ERK activation and LTC4 production in basophils but has little effect on histamine release or cytokine synthesis (IL-4 and IL-13).107,116 The above studies highlight that ERK activation does not seem to affect cytokine transcription in basophils although it is an important regulator of cJun, cFos, STAT, and other transcription factors in diverse cell types. Therefore, in basophils ERKs appear to be largely involved only in LTC4 production because of the activation of PLA2. Nevertheless, we have also demonstrated that ERK activation is not by itself sufficient for leukotriene synthesis because IL-3, which strongly activates ERK in the absence of IgE-dependent stimulation,116 is almost ineffective at causing LTC4release.74,107,117,118 Despite this, in combination with anti-IgE, IL-3 leads to both a striking potentiation and prolongation of ERK phosphorylation compared with the effects of IL-3 or anti-IgE alone and leads to twice as much LTC4 production than anti-IgE alone. Although activation of PLA2 is dependent on ERK, translocation of soluble PLA2 from the cytosol to the membrane is dependent on calcium. However, studies by Krieger et al119 and MacGlashan and Hubbard118 have shown that the priming caused by briefly (< 15 minutes) preincubating basophils with IL-3 neither requires extracellular calcium nor induces calcium elevations. Taken together, these findings strongly suggest that, despite there being an important regulatory input from both ERK phosphorylation and intracellular calcium mobilization regarding PLA2 activity, one or more additional signals, possibly from protein kinase C (PKC)–dependent routes,120 are vital for LTC4 synthesis in basophils.

Comparing human basophils with other rodent histamine-producing cells, there are important differences with respect to regulation of cytokine synthesis controlled through ERKs. For example, IgE-dependent TNF-α production in RBL-2H3 cells is inhibited by the MEK inhibitor PD-098059, suggesting ERK involvement. In contrast, the release of IgE-dependent TNF-α from MC/9 murine mast cells is not affected by the antagonist. Although basophils have not been shown to produce TNF-α, the above examples highlight further the variable abilities of ERKs to control the transcription of cytokine factors between human basophils and their counterparts in rodents. Further evidence that the MEK-ERK route does not predominantly influence basophil IL-4 production is provided by Miura et al,116 who report that the bacterial peptide fMLP induces basophil ERK activation without IL-4 release.

Calcium dependence of basophil mediator secretion

It has long been appreciated that basophil degranulation is strikingly reduced in the absence of extracellular calcium.121 Similarly, production of both IL-4 113,122,123 and IL-13 (B.F.G. et al, unpublished data, 1998) is just as dependent on the ion for optimum response to IgE-receptor activation. Calcium responses are in 2 phases.124 The first takes place by the release of intracellular calcium stores from the endoplasmic reticulum because of the effects of inositol 1,4,5-trisphosphate (IP3). The second phase is caused by the opening of calcium channels allowing an influx of the ion from the extracellular milieu and is modulated by cyclic adenosine monophosphate (cAMP)–elevating drugs.125A rise in intracellular free calcium ions can be mimicked by using calcium ionophores such as A23187, which directly transport extracellular calcium into the cells, release internal stores of the ion, and evoke degranulation, eicosanoid generation, and cytokine production.

In terms of IgE-dependent cytokine secretion, both IL-4 and IL-13 are controlled by calcium-calcineurin pathways, which promote the translocation of the transcription factor NFAT to the nucleus.126 This pathway is blocked by macrolides such as FK506 and cyclosporin A, which reduce both IL-4 and IL-13 synthesis.127 Additionally, the release of these cytokines is also effectively blocked by cAMP-elevating drugs such as theophylline or β-2 agonists (salmeterol), further highlighting the importance of calcium influx in basophil activation.128 

IL-4 and IL-13 are differentially controlled by IL-3 and PKC

In terms of IgE-dependent stimulation of IL-4 and IL-13, there is no clear evidence of differential control regarding the production of these cytokines. However, the kinetics of their release from basophils is strikingly different, suggesting that factors other than the signaling routes already described above are involved. IL-4 is released rapidly, peaking within 4 hours of stimulation, and is accompanied in some donors by the release of preformed stores of this cytokine.6,129,130 In contrast, IL-13 is detectable in basophil supernatants beginning 2 hours after FcεRI cross-linking, and the amounts of this factor continue to rise for at least 16 hours.6 At present, the reasons for these differing kinetics are unknown, but factors such as autocrine effects are now being investigated.

In terms of the levels of IL-4 and IL-13 produced, there is growing evidence that signals employed by priming factors such as IL-3 favor the production of IL-13 rather than IL-4. IL-3 potentiates the IgE-dependent release of both factors but, in addition to this, Schroeder et al, as well as Redrup et al, have reported that IL-3 stimulation alone is sufficient itself to give rise to IL-13 generation without a marked increase in IL-4 production.126,127 These authors further showed that, in stark contrast to IgE signaling, this IL-3-dependent IL-13 release is insensitive to FK506, suggesting the involvement of an activatory pathway independent of calcineurin and NFAT.

A further tier of differential control of IL-4 and IL-13 release is seen with respect to PKC. Following PMA-induced PKC activation in basophils, Schroeder and coworkers have reported that although messenger RNA expressions for both IL-4 and IL-13 are increased only IL-13 protein is released.126,127 It is not yet clear, however, which isozymes of PKC are involved. The same authors have shown that the selective PKC inhibitor bisindolylmaleimide II not only reverses the effects of PMA activation but potentiates the anti-IgE induced IL-4 release but not that of IL-13. Interestingly, bisindolylmaleimide II, as well as FK506, does not affect IL-3–induced secretion of IL-13, thus showing that IL-3–associated signaling in basophils diverges considerably from both calcium and PKC-sensitive routes compared with IgE-dependent pathways.

Future aspects

The brief summary above regarding the control of IgE signaling in basophils highlights 2 major problems for future consideration. First, functional differences exist between primary basophils and mast cells or basophil cell lines and, second, IgE-dependent signaling is greatly affected by the actions of priming factors such as IL-3. Thus, the net outcome of IgE-triggered basophil activation in vivo is likely to be modulated by varying degrees of the local concentrations of hematopoietic cytokines (such as IL-3, IL-5, and GM-CSF) as well as neurotrophic cytokines (NGF), complement factors, and histamine-releasing factors. Because basophils are geared to the rapid release of Th2-type cytokines, it would be of considerable interest to establish the concentrations of the above factors in the in vivo environment, particularly during an allergic reaction where acute (IgE-mediated) basophil activation occurs.

Although their existence has been known for more than a century, the physiologic role of basophils in immunity remains a mystery. It is conceivable that basophils, like eosinophils, neutrophils, macrophages, and mast cells, fulfill important roles in innate immunity against pathogenic organisms. Important clues could be derived from the study of natural or experimentally induced gene deficiencies, as happened with W/Wv c-kit–deficient mice,131 which have proven very useful in dissecting the role of mast cells in immunity to intestinal nematodes, or IL-3−/− mice.132Unfortunately, in the absence of a precise knowledge about the unique developmental pathway of basophils, none of the currently available gene-deficient mice are likely to elucidate the exclusive role of basophils in immunity.

Mast cells and basophils have been proposed to play an important role in the innate immune response to a variety of pathogens.133 For mast cells such a role is corroborated by an increasing body of evidence,133,134 whereas the current evidence for a comparable involvement of basophils is, at best, circumstantial. Such a role would require the ability of basophils to be activated in a non–antigen-specific manner. Indeed, several such non–antigen-specific stimuli, derived from various organisms, have been shown to induce mediator or cytokine release from basophils. For example, protein Fv, an endogenous Ig-binding protein released in the intestine of patients affected by viral hepatitis, as well as the HIV-1 glycoprotein gp 120 have recently been shown to induce IL-4 and IL-13 production from basophils and mast cells by binding to the VH3 region of IgE.135-137 Similarly, antigens derived from the egg stage of the parasite Schistosoma mansoni (SEA, or soluble egg antigen) induce the release of IL-4 and other mediators from basophils of nonimmune donors.138This activation was dependent on the presence of IgE because a short incubation at low pH (under conditions that induce dissociation of IgE from its receptor)139 abrogated, whereas resensitization with purified IgE or other IgE-containing materials restored the activating effect of SEA (Falcone et al138 and K. Haisch et al, unpublished data, 2000). Furthermore, proteases secreted by the hookworm Necator americanus induce the de novo synthesis of IL-4 and IL-13 in the basophilic cell line KU812 (C. Phillips et al, unpublished data, 1999). The filarial parasiteBrugia malayi strongly expresses the geneBm-tph-1,140 which is closely related to a human IgE-dependent histamine releasing factor,141 but it is not known whether Bm-TPH-1 can also activate basophils. Finally, the recombinant form of the IgE-dependent human histamine-releasing factor induces histamine and IL-4 release from basophils. This factor, in contrast to original findings,142,143 has recently been shown to act independently of the presence of IgE and is now thought to bind to a receptor present on the surface of basophils.144 

Because basophils rapidly release considerable amounts of IL-4 upon stimulation, they may have a decisive impact on the outcome of a primary infection by inducing T-cell differentiation to the Th2 phenotype. To the best of our knowledge, basophils are the only leukocytes containing preformed IL-4, although in relatively small amounts and not detected in all donors. Constitutive IL-4 expression in unstimulated basophils has been demonstrated by intracellular cytokine staining in saponin-permeabilized cells130 as well as by enzyme-linked immunosorbent assay.6 Apart from basophils, cells such as Th2 cells, NK1.1+ cells, γδ+T cells, eosinophils, and mast cells also have been found to produce IL-4 and have been considered as a cellular source of the so-called “early IL-4.”145 The discussion about the in vivo source of early IL-4 has led to much controversy146; the bottom line is that several different cell types may play a not mutually exclusive role in initiating Th2 responses.145,146 It is also possible that part of the controversy is ultimately due to fundamental differences in the relative roles of mast cells and basophils as a source of IL-4 in rodents versus humans.

Another line of evidence suggesting a possible role of basophils as a source of early IL-4 comes from the study of the effects of plant lectins on this cell type. It has long been known that lectins such as concanavalin A can trigger degranulation and histamine release from basophils.147,148 Using a panel of 16 plant lectins with different carbohydrate specificities, we have recently shown that lectins differentially induce the release of IL-4 (and IL-13) from basophils.149 The capacity of the studied lectins to trigger cytokine release correlated with their binding to different myeloma IgE molecules—IgE-PS, IgE-(T+H), IgE-PE, IgE-VL—suggesting non–antigen-specific cross-linking of FcεRI-bound IgE via its carbohydrate moieties as the initial step of signal generation.149 However, when unsensitized RBL-2H3 cells, ie, cells devoid of receptor-bound IgE, were stimulated, most of the cytokine-inducing lectins also triggered the release of β-hexosaminidase (a granule mediator in rodent mast cells). OnlySambucus nigra and Ricinus communis agglutinins clearly required sensitization of the RBL-2H3 cells with rat IgE to elicit a full response (H. Haas et al, unpublished data, 1999). By analogy, a similar IgE-independent mechanism may be expected for IL-4 release induced by most lectins. Although the significance of these in vitro findings needs to be confirmed in vivo, it nevertheless points to a whole category of molecules with the ability to trigger the release of IL-4 from basophils in a non–antigen-specific manner. It is therefore intriguing to note that several parasitic helminths, which are well known to induce a strong Th2 response in their host, have been recently shown to produce or secrete lectins.150 For instance, infective larvae of the nematode parasite Toxocara canis secrete a novel C-type lectin (TES-32) that surprisingly binds both mannose- and galactose-type monosaccharides,151 which are both present on the carbohydrate side chains of IgE.152,153 TES-32 is the most abundant glycoprotein secreted by this parasite in vitro. The hematophagous parasite N americanus produces a major allergen that is a putative homolog of calreticulin,154 a lectin with a carbohydrate affinity155 that also matches the glycosylation pattern of human IgE. Thus, several pathogenic organisms may induce the activation of basophils by the release of superallergens, a term introduced by Patella et al,135,136which in this context would define molecules with the ability to cross-link FcεRI either directly or via receptor-bound IgE in a non–antigen-specific manner.

Taken together, the observation that different classes of molecules derived from pathogens or endogenous sources can directly activate basophils independently of the presence of specific IgE on their surface fulfills the theoretical requirement for a role of this cell type in innate immunity or in bridging innate and specific immunity by skewing the differentiation of naive T cells to the Th2 phenotype. Thus, more research is needed to address the fundamental question concerning the true physiologic role of basophils in the immune system.

Investigation into the role of basophils in health and disease has previously been curtailed by the lack of suitable experimental tools. The following section, therefore, summarizes the currently available tools for basophil research.

Monoclonal antibodies

Because basophils are quickly recruited to the tissues in a variety of allergic diseases, monoclonal antibodies allowing specific detection of basophils by immunohistochemical staining would be a highly desirable tool, eg, for studying the relative contribution of this cell type to pathogenesis. Earlier protocols relied on the presence of FcεRI and the absence of tryptase for immunohistochemical detection.156,157 It has become clear, however, that the expression of FcεRI is not restricted to mast cells and basophils, because it can be up-regulated on other cells such as Langerhans cells158 and other dendritic cells,159eosinophils,160 and monocytes.161Furthermore, the expression of tryptase cannot be used for discrimination of mast cells from basophils, because the gene for tryptase α is transcribed in the latter cell type162 and is present in substantial amounts in peripheral blood basophils of patients with asthma or other allergic diseases.20Therefore, several laboratories have generated antibodies for specific detection of basophils in flow cytometry or immunohistochemistry.

There are currently 3 basophil-specific monoclonal antibodies: BSP-1, 2D7, and BB-1. BSP-1 is an IgM class monoclonal antibody that recognizes an epitope expressed on the surface of human basophils and was originally raised against the human erythroblastic leukemia cell line (HEL).163 It reacts with a 45-kd surface antigen on HEL cells in Western blots.163 Because of its low sensitivity, BSP-1 is not suitable for immunohistochemistry.164 Monoclonal antibody 2D7 is an IgG1κ monoclonal antibody that reacts with a ligand localized to the secretory granules of human basophils.164 Western blots with basophil extracts exhibit 2 major bands with apparent molecular weights of 72 and 76 kd and a minor band of 124 kd.164Immunologic activation of basophils leads to the loss or attenuation of the staining intensity, indicating release (or decay) of the 2D7 antigen.164 Monoclonal antibody 2D7 has successfully been used for immunohistochemical staining of basophils in human skin during the LPR after cutaneous allergen challenge.165 

The most recent addition to the pharmacopoeia is BB-1, an IgG2a monoclonal antibody that recognizes a 124- ( ± 11) kd antigen (estimated by the method of Hedrick and Smith)166 localized mainly to the secretory granules with a comparatively small expression on the surface.167 The BB-1 antigen was released upon activation with anti-IgE or the calcium ionophoreA23187.167 Because the BB-1 antigen was not detected in any other cell types, the authors suggest its use as a discriminating marker of basophil activation, particularly in the tissues, where it has been successfully used for immunohistochemical detection of basophils in lung sections fixed in Carnoy's fluid.167 

Cell lines

Several studies have used the cell line KU812, originally established from a patient with chronic myelogenous leukemia.168 KU812 is seen as an immature basophil precursor169 and has served as a model for basophil differentiation. Several cytokines can induce its differentiation into basophil-like cells, including TNF-α and IL-6,170,171IL-3,172 and IL-4.173 As shown by Hara et al,173 incubation of KU812 cells with IL-4 induced several changes, including a 10-fold increase of total histamine content, appearance of metachromatic staining with toluidine blue, and up-regulation of functionally active FcεRI on the surface after 7 days, with increased transcription of FcεRI α, β, and γ chains' messenger RNA. Another cell line, termed LAMA-84, was established in 1987 by Seigneurin et al174 and has also been described as basophil-like.175 Kepley and coworkers176 have recently described the in vitro generation of fully functional basophils from cord blood. The protocol consists in pulsing normal cord blood leukocytes with IL-3 for 3 to 4 hours and subsequently incubating them with fetal calf serum. The resulting cells are mostly 2D7+, FcεRI+, express a series of integrins also found on normal peripheral blood basophils (CD11b, CD18, CD29, and CD49d), and display basophil-like morphology by light or electron microscopy. Activation through the high-affinity IgE receptor results in a time- and dose-dependent release of histamine. The use of basophil-like cell lines, however, must be cautiously examined given the fundamental differences in morphology and function compared with primary human basophils.

Purification protocols

A high degree of basophil purity is often required for functional studies with basophils, especially when mediator secretion from contaminating cells masks what is released by basophils. Pure basophil populations are also mandatory when investigating either intracellular signaling events or to exclude the possibility of priming cytokines (eg, IL-3, GM-CSF), derived from cellular contaminants, to influence basophil function. Therefore, several protocols for basophil purification have been published over the last decade (see references in Haisch et al177). When purifying basophils, one is faced with several challenges. First, the average percentage of basophils in the peripheral blood of healthy individuals is low (< 1%), which restricts the possible yield. Furthermore, a large donor variation in the proportion of basophils makes the final recovery of basophils unpredictable, especially when dealing with blood from unknown donors (ie, from buffy coats).178 

Most of the currently published purification techniques rely on initial physical separation methods (density gradients or elutriation) that, by themselves, do not reliably result in very high purity but are successful in enriching basophils sufficiently enough for further purification by immunoselection. Some protocols have relied on the expression of the high-affinity receptor FcεRI on the surface of human basophils.179,180 This positive selection technique, although resulting in very high basophil purity, is not desirable for many purposes because of the likelihood of activation during purification. The use of negative selection with magnetic beads176,181,182 consistently resulted in viable, functional (nonpreactivated) basophils but, in our experience, high yields were usually obtained only with hyperbasophilic donors.178 A recently published protocol, however, has overcome this limitation (Figure 3) and uses a commercially available kit,177 therefore making the purification protocol widely accessible to any laboratory with an interest in granulocyte function. A similar protocol combines density gradient centrifugation with negative selection with magnetic beads.183 

Fig. 3.

Purified human peripheral blood basophils stained with May-Grünwald-Giemsa obtained with a recently published basophil purification protocol.177

(A) Original magnification × 160. (B) Original magnification × 1000. Courtesy of A. Gronow and K. Haisch, Forschungszentrum Borstel, Germany.

Fig. 3.

Purified human peripheral blood basophils stained with May-Grünwald-Giemsa obtained with a recently published basophil purification protocol.177

(A) Original magnification × 160. (B) Original magnification × 1000. Courtesy of A. Gronow and K. Haisch, Forschungszentrum Borstel, Germany.

Close modal

Taken together, the availability of effective protocols for the purification of nearly homogeneous basophils or for the generation of this cell type from peripheral blood precursors as well as the availability of monoclonal antibodies for immunohistochemical detection and of basophil-like cell lines should encourage further study of human basophil function. The development of similar tools for the study of murine basophils would be extremely useful because it would enable the research to be extended to experimental models of disease.

The authors thank Dr Martin J. Holland for critical comments and suggestions.

Supported by the Deutsche Forschungsgemeinschaft (Ha 1590/2-1 and Fa 359/1-1).

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Ehrlich
P
Beiträge zur Kenntnis der granulierten Bindegewebszellen und der eosinophilen Leukocythen.
Arch Anat Physiol.
1879
166
169
2
Ehrlich P. Beiträge zur Theorie und Praxis der histologischen Färbung [thesis]. Leipzig: 1878.
3
Brunner
T
Heusser
CH
Dahinden
CA
Human peripheral blood basophils primed by interleukin 3 (IL-3) produce IL-4 in response to immunoglobulin E receptor stimulation.
J Exp Med.
177
1993
605
611
4
Arock
M
Merle-Beral
H
Dugas
B
et al
IL-4 release by human leukemic and activated normal basophils.
J Immunol.
151
1993
1441
1447
5
Li
H
Sim
TC
Alam
R
IL-13 released by and localized in human basophils.
J Immunol.
156
1996
4833
4838
6
Gibbs
BF
Haas
H
Falcone
FH
et al
Purified human peripheral blood basophils release interleukin-13 and preformed interleukin-4 following immunological activation.
Eur J Immunol.
26
1996
2493
2498
7
Ochensberger
B
Daepp
GC
Rihs
S
Dahinden
CA
Human blood basophils produce interleukin-13 in response to IgE-receptor-dependent and -independent activation.
Blood.
88
1996
3028
3037
8
Yanagihara
Y
Kajiwara
K
Basaki
Y
et al
Cultured basophils but not cultured mast cells induce human IgE synthesis in B cells after immunologic stimulation.
Clin Exp Immunol.
111
1998
136
143
9
Gauchat
JF
Henchoz
S
Mazzei
G
et al
Induction of human IgE synthesis in B cells by mast cells and basophils.
Nature.
365
1993
340
343
10
Uguccioni
M
Mackay
CR
Ochensberger
B
et al
High expression of the chemokine receptor CCR3 in human blood basophils. Role in activation by eotaxin, MCP-4, and other chemokines.
J Clin Invest.
100
1997
1137
1143
11
Valent
P
The phenotype of human eosinophils, basophils, and mast cells.
J Allergy Clin Immunol.
94(suppl)
1994
1177
1183
12
Valent
P
Immunophenotypic characterization of human basophils and mast cells.
Chem Immunol.
61
1995
34
48
13
Boyce
JA
Friend
D
Matsumoto
R
Austen
KF
Owen
WF
Differentiation in vitro of hybrid eosinophil/basophil granulocytes: autocrine function of an eosinophil developmental intermediate.
J Exp Med.
182
1995
49
57
14
Leary
AG
Ogawa
M
Identification of pure and mixed basophil colonies in culture of human peripheral blood and marrow cells.
Blood.
64
1984
78
83
15
Denburg
JA
Telizyn
S
Messner
H
et al
Heterogeneity of human peripheral blood eosinophil-type colonies: evidence for a common basophil-eosinophil progenitor.
Blood.
66
1985
312
318
16
Weil
SC
Hrisinko
MA
A hybrid eosinophilic-basophilic granulocyte in chronic granulocytic leukemia.
Am J Clin Pathol.
87
1987
66
70
17
Kempuraj
D
Saito
H
Kaneko
A
et al
Characterization of mast cell-committed progenitors present in human umbilical cord blood.
Blood.
93
1999
3338
3346
18
Kirshenbaum
AS
Goff
JP
Semere
T
Foster
B
Scott
LM
Metcalfe
DD
Demonstration that human mast cells arise from a progenitor cell population that is CD34(+), c-kit(+), and expresses aminopeptidase N (CD13).
Blood.
94
1999
2333
2342
19
Buhring
HJ
Simmons
PJ
Pudney
M
et al
The monoclonal antibody 97A6 defines a novel surface antigen expressed on human basophils and their multipotent and unipotent progenitors.
Blood.
94
1999
2343
2356
20
Li
L
Li
Y
Reddel
SW
et al
Identification of basophilic cells that express mast cell granule proteases in the peripheral blood of asthma, allergy, and drug-reactive patients.
J Immunol.
161
1998
5079
5086
21
Agis
H
Willheim
M
Sperr
WR
et al
Monocytes do not make mast cells when cultured in the presence of SCF. Characterization of the circulating mast cell progenitor as a c-kit+, CD34+, Ly-, CD14-, CD17-, colony-forming cell.
J Immunol.
151
1993
4221
4227
22
Agis
H
Beil
WJ
Bankl
HC
et al
Mast cell-lineage versus basophil lineage involvement in myeloproliferative and myelodysplastic syndromes: diagnostic role of cell-immunophenotyping.
Leuk Lymphoma.
22
1996
187
204
23
Kishi
K
Takahashi
M
Aoki
S
et al
[A new Ph1 positive cell line (KU812) from a patient with blastic crisis of chronic myelogenous leukemia].
Nippon Ketsueki Gakkai Zasshi.
47
1984
709
718
24
Kirshenbaum
AS
Goff
JP
Kessler
SW
Mican
JM
Zsebo
KM
Metcalfe
DD
Effect of IL-3 and stem cell factor on the appearance of human basophils and mast cells from CD34+ pluripotent progenitor cells.
J Immunol.
148
1992
772
777
25
Saito
H
Hatake
K
Dvorak
AM
et al
Selective differentiation and proliferation of hematopoietic cells induced by recombinant human interleukins.
Proc Natl Acad Sci U S A.
85
1988
2288
2292
26
Denburg
JA
Silver
JE
Abrams
JS
Interleukin-5 is a human basophilopoietin: induction of histamine content and basophilic differentiation of HL-60 cells and of peripheral blood basophil-eosinophil progenitors.
Blood.
77
1991
1462
1468
27
Yamaguchi
M
Hirai
K
Morita
Y
et al
Hemopoietic growth factors regulate the survival of human basophils in vitro.
Int Arch Allergy Immunol.
97
1992
322
329
28
Tsuda
T
Wong
D
Dolovich
J
Bienenstock
J
Marshall
J
Denburg
JA
Synergistic effects of nerve growth factor and granulocyte-macrophage colony-stimulating factor on human basophilic cell differentiation.
Blood.
77
1991
971
979
29
Valent
P
Schmidt
G
Besemer
J
et al
Interleukin-3 is a differentiation factor for human basophils.
Blood.
73
1989
1763
1769
30
Iemura
A
Tsai
M
Ando
A
Wershil
BK
Galli
SJ
The c-kit ligand, stem cell factor, promotes mast cell survival by suppressing apoptosis.
Am J Pathol.
144
1994
321
328
31
Galli
SJ
New concepts about the mast cell.
N Engl J Med.
328
1993
257
265
32
Thomas
LL
Basophil and eosinophil interactions in health and disease.
Chem Immunol.
61
1995
186
207
33
Knol
EF
Mul
FP
Lie
WJ
Verhoeven
AJ
Roos
D
The role of basophils in allergic disease.
Eur Respir J Suppl.
22
1996
126s
131s
34
Naclerio
RM
Proud
D
Togias
AG
et al
Inflammatory mediators in late antigen-induced rhinitis.
N Engl J Med.
313
1985
65
70
35
Charlesworth
EN
Hood
AF
Soter
NA
Kagey-Sobotka
A
Norman
PS
Lichtenstein
LM
Cutaneous late-phase response to allergen. Mediator release and inflammatory cell infiltration.
J Clin Invest.
83
1989
1519
1526
36
Liu
MC
Hubbard
WC
Proud
D
et al
Immediate and late inflammatory responses to ragweed antigen challenge of the peripheral airways in allergic asthmatics. Cellular, mediator, and permeability changes.
Am Rev Respir Dis.
144
1991
51
58
37
Bascom
R
Wachs
M
Naclerio
RM
Pipkorn
U
Galli
SJ
Lichtenstein
LM
Basophil influx occurs after nasal antigen challenge: effects of topical corticosteroid pretreatment.
J Allergy Clin Immunol.
81
1988
580
589
38
Rothenberg
ME
Zimmermann
N
Mishra
A
et al
Chemokines and chemokine receptors: their role in allergic airway disease.
J Clin Immunol.
19
1999
250
265
39
Nickel
R
Beck
LA
Stellato
C
Schleimer
RP
Chemokines and allergic disease.
J Allergy Clin Immunol.
104
1999
723
742
40
Homey
B
Zlotnik
A
Chemokines in allergy.
Curr Opin Immunol.
11
1999
626
634
41
Bochner
BS
Sterbinsky
SA
Briskin
M
Saini
SS
MacGlashan
DW
Jr
Counter-receptors on human basophils for endothelial cell adhesion molecules.
J Immunol.
157
1996
844
850
42
Schleimer
RP
Sterbinsky
SA
Kaiser
J
et al
IL-4 induces adherence of human eosinophils and basophils but not neutrophils to endothelium. Association with expression of VCAM-1.
J Immunol.
148
1992
1086
1092
43
Bochner
BS
Luscinskas
FW
Gimbrone
MA
Jr
et al
Adhesion of human basophils, eosinophils, and neutrophils to interleukin 1-activated human vascular endothelial cells: contributions of endothelial cell adhesion molecules.
J Exp Med.
173
1991
1553
1557
44
Corrigan
CJ
Eotaxin and asthma: some answers, more questions.
Clin Exp Immunol.
116
1999
1
3
45
Jose
PJ
Griffiths-Johnson
DA
Collins
PD
et al
Eotaxin: a potent eosinophil chemoattractant cytokine detected in a guinea pig model of allergic airways inflammation.
J Exp Med.
179
1994
881
887
46
Rothenberg
ME
Luster
AD
Lilly
CM
Drazen
JM
Leder
P
Constitutive and allergen-induced expression of eotaxin mRNA in the guinea pig lung.
J Exp Med.
181
1995
1211
1216
47
Ganzalo
JA
Jia
GQ
Aguirre
V
et al
Mouse Eotaxin expression parallels eosinophil accumulation during lung allergic inflammation but it is not restricted to a Th2-type response.
Immunity.
4
1996
1
14
48
Griffiths-Johnson
DA
Collins
PD
Jose
PJ
Williams
TJ
Animal models of asthma: role of chemokines.
Methods Enzymol.
288
1997
241
266
49
Humbles
AA
Conroy
DM
Marleau
S
et al
Kinetics of eotaxin generation and its relationship to eosinophil accumulation in allergic airways disease: analysis in a guinea pig model in vivo.
J Exp Med.
186
1997
601
612
50
Ying
S
Robinson
DS
Meng
Q
et al
Enhanced expression of eotaxin and CCR3 mRNA and protein in atopic asthma. Association with airway hyperresponsiveness and predominant co-localization of eotaxin mRNA to bronchial epithelial and endothelial cells.
Eur J Immunol.
27
1997
3507
3516
51
Lamkhioued
B
Renzi
PM
Abi-Younes
S
et al
Increased expression of eotaxin in bronchoalveolar lavage and airways of asthmatics contributes to the chemotaxis of eosinophils to the site of inflammation.
J Immunol.
159
1997
4593
4601
52
Mattoli
S
Stacey
MA
Sun
G
Bellini
A
Marini
M
Eotaxin expression and eosinophilic inflammation in asthma.
Biochem Biophys Res Commun.
236
1997
299
301
53
Brown
JR
Kleimberg
J
Marini
M
Sun
G
Bellini
A
Mattoli
S
Kinetics of eotaxin expression and its relationship to eosinophil accumulation and activation in bronchial biopsies and bronchoalveolar lavage (BAL) of asthmatic patients after allergen inhalation.
Clin Exp Immunol.
114
1998
137
146
54
Rothenberg
ME
MacLean
JA
Pearlman
E
Luster
AD
Leder
P
Targeted disruption of the chemokine eotaxin partially reduces antigen-induced tissue eosinophilia.
J Exp Med.
185
1997
785
790
55
Yamada
H
Hirai
K
Miyamasu
M
et al
Eotaxin is a potent chemotaxin for human basophils.
Biochem Biophys Res Commun.
231
1997
365
368
56
Dahinden
CA
Geiser
T
Brunner
T
et al
Monocyte chemotactic protein 3 is a most effective basophil- and eosinophil-activating chemokine.
J Exp Med.
179
1994
751
756
57
Garcia-Zepeda
EA
Combadiere
C
Rothenberg
ME
et al
Human monocyte chemoattractant protein (MCP)-4 is a novel CC chemokine with activities on monocytes, eosinophils, and basophils induced in allergic and nonallergic inflammation that signals through the CC chemokine receptors (CCR)-2 and -3.
J Immunol.
157
1996
5613
5626
58
Lukacs
NW
Standiford
TJ
Chensue
SW
Kunkel
RG
Strieter
RM
Kunkel
SL
C-C chemokine-induced eosinophil chemotaxis during allergic airway inflammation.
J Leukoc Biol.
60
1996
573
578
59
Lukacs
NW
Strieter
RM
Warmington
K
Lincoln
P
Chensue
SW
Kunkel
SL
Differential recruitment of leukocyte populations and alteration of airway hyperreactivity by C-C family chemokines in allergic airway inflammation.
J Immunol.
158
1997
4398
4404
60
Gonzalo
JA
Lloyd
CM
Wen
D
et al
The coordinated action of CC chemokines in the lung orchestrates allergic inflammation and airway hyperresponsiveness.
J Exp Med.
188
1998
157
167
61
Kitaura
M
Suzuki
N
Imai
T
et al
Molecular cloning of a novel human CC chemokine (Eotaxin-3) that is a functional ligand of CC chemokine receptor 3.
J Biol Chem.
274
1999
27975
27980
62
Shinkai
A
Yoshisue
H
Koike
M
et al
A novel human CC chemokine, eotaxin-3, which is expressed in IL-4-stimulated vascular endothelial cells, exhibits potent activity toward eosinophils.
J Immunol.
163
1999
1602
1610
63
Iliopoulos
O
Baroody
FM
Naclerio
RM
Bochner
BS
Kagey-Sobotka
A
Lichtenstein
LM
Histamine-containing cells obtained from the nose hours after antigen challenge have functional and phenotypic characteristics of basophils.
J Immunol.
148
1992
2223
2228
64
Kay
AB
Ying
S
Varney
V
et al
Messenger RNA expression of the cytokine gene cluster, interleukin 3 (IL-3), IL-4, IL-5, and granulocyte/macrophage colony-stimulating factor, in allergen-induced late-phase cutaneous reactions in atopic subjects.
J Exp Med.
173
1991
775
778
65
Yamaguchi
M
Hirai
K
Shoji
S
et al
Haemopoietic growth factors induce human basophil migration in vitro.
Clin Exp Allergy.
22
1992
379
383
66
Tanimoto
Y
Takahashi
K
Takata
M
Kawata
N
Kimura
I
Purification of human blood basophils using negative selection by flow cytometry.
Clin Exp Allergy.
22
1992
1015
1019
67
Hirai
K
Morita
Y
Miyamoto
T
Hemopoietic growth factors regulate basophil function and viability.
Immunol Ser.
57
1992
587
600
68
Bischoff
SC
Dahinden
CA
Effect of nerve growth factor on the release of inflammatory mediators by mature human basophils.
Blood.
79
1992
2662
2669
69
Sanico
AM
Koliatsos
VE
Stanisz
AM
Bienenstock
J
Togias
A
Neural hyperresponsiveness and nerve growth factor in allergic rhinitis.
Int Arch Allergy Immunol.
118
1999
154
158
70
Bonini
S
Lambiase
A
Angelucci
F
Magrini
L
Manni
L
Aloe
L
Circulating nerve growth factor levels are increased in humans with allergic diseases and asthma.
Proc Natl Acad Sci U S A.
93
1996
10955
10960
71
Burgi
B
Brunner
T
Dahinden
CA
The degradation product of the C5a anaphylatoxin C5adesarg retains basophil-activating properties.
Eur J Immunol.
24
1994
1583
1589
72
MacDonald
SM
Schleimer
RP
Kagey-Sobotka
A
Gillis
S
Lichtenstein
LM
Recombinant IL-3 induces histamine release from human basophils.
J Immunol.
142
1989
3527
3532
73
Kuna
P
Reddigari
SR
Kornfeld
D
Kaplan
AP
IL-8 inhibits histamine release from human basophils induced by histamine-releasing factors, connective tissue activating peptide III, and IL-3.
J Immunol.
147
1991
1920
1924
74
Kurimoto
Y
de Weck
AL
Dahinden
CA
Interleukin 3-dependent mediator release in basophils triggered by C5a.
J Exp Med.
170
1989
467
479
75
Bischoff
SC
de Weck
AL
Dahinden
CA
Interleukin 3 and granulocyte/macrophage-colony-stimulating factor render human basophils responsive to low concentrations of complement component C3a.
Proc Natl Acad Sci U S A.
87
1990
6813
6817
76
Brunner
T
de Weck
AL
Dahinden
CA
Platelet-activating factor induces mediator release by human basophils primed with IL-3, granulocyte-macrophage colony-stimulating factor, or IL-5.
J Immunol.
147
1991
237
242
77
Sarmiento
EU
Espiritu
BR
Gleich
GJ
Thomas
LL
IL-3, IL-5, and granulocyte-macrophage colony-stimulating factor potentiate basophil mediator release stimulated by eosinophil granule major basic protein.
J Immunol.
155
1995
2211
2221
78
Ochensberger
B
Tassera
L
Bifrare
D
Rihs
S
Dahinden
CA
Regulation of cytokine expression and leukotriene formation in human basophils by growth factors, chemokines and chemotactic agonists.
Eur J Immunol.
29
1999
11
22
79
Dvorak
AM
Lett-Brown
M
Thueson
D
Grant
JA
Complement-induced degranulation of human basophils.
J Immunol.
126
1981
523
528
80
Fureder
W
Agis
H
Willheim
M
et al
Differential expression of complement receptors on human basophils and mast cells. Evidence for mast cell heterogeneity and CD88/C5aR expression on skin mast cells.
J Immunol.
155
1995
3152
3160
81
Agis
H
Fureder
W
Bankl
HC
et al
Comparative immunophenotypic analysis of human mast cells, blood basophils and monocytes.
Immunology.
87
1996
535
543
82
van Gils
FC
van Teeffelen
ME
Neelis
KJ
et al
Interleukin-3 treatment of rhesus monkeys leads to increased production of histamine-releasing cells that express interleukin-3 receptors at high levels.
Blood.
86
1995
592
597
83
van Gils
FC
Mulder
AH
van den Bos
C
Burger
H
van Leen
RW
Wagemaker
G
Acute side effects of homologous interleukin-3 in rhesus monkeys.
Am J Pathol.
143
1993
1621
1633
84
Kasaian
MT
Clay
MJ
Happ
MP
Garman
RD
Hirani
S
Luqman
M
IL-4 production by allergen-stimulated primary cultures: identification of basophils as the major IL-4-producing cell type.
Int Immunol.
8
1996
1287
1297
85
Devouassoux
G
Foster
B
Scott
LM
Metcalfe
DD
Prussin
C
Frequency and characterization of antigen-specific IL-4- and IL-13- producing basophils and T cells in peripheral blood of healthy and asthmatic subjects.
J Allergy Clin Immunol.
104
1999
811
819
86
Moser
R
Groscurth
P
Carballido
JM
et al
Interleukin-4 induces tissue eosinophilia in mice: correlation with its in vitro capacity to stimulate the endothelial cell-dependent selective transmigration of human eosinophils.
J Lab Clin Med.
122
1993
567
575
87
Sanz
MJ
Marinova-Mutafchieva
L
Green
P
Lobb
RR
Feldmann
M
Nourshargh
S
IL-4-induced eosinophil accumulation in rat skin is dependent on endogenous TNF-alpha and alpha 4 integrin/VCAM-1 adhesion pathways.
J Immunol.
160
1998
5637
5645
88
Rankin
JA
Picarella
DE
Geba
GP
et al
Phenotypic and physiologic characterization of transgenic mice expressing interleukin 4 in the lung: lymphocytic and eosinophilic inflammation without airway hyperreactivity.
Proc Natl Acad Sci U S A.
93
1996
7821
7825
89
Hickey
MJ
Granger
DN
Kubes
P
Molecular mechanisms underlying IL-4-induced leukocyte recruitment in vivo: a critical role for the alpha4.
J Immunol.
163
1999
3441
3448
90
Mochizuki
M
Bartels
J
Mallet
AI
Christophers
E
Schroder
JM
IL-4 induces eotaxin: a possible mechanism of selective eosinophil recruitment in helminth infection and atopy.
J Immunol.
160
1998
60
68
91
Devouassoux
G
Metcalfe
DD
Prussin
C
Eotaxin potentiates antigen-dependent basophil IL-4 production.
J Immunol.
163
1999
2877
2882
92
Kroegel
C
Julius
P
Matthys
H
Virchow
JC
Jr
Luttmann
W
Endobronchial secretion of interleukin-13 following local allergen challenge in atopic asthma: relationship to interleukin-4 and eosinophil counts.
Eur Respir J.
9
1996
899
904
93
Wills-Karp
M
Luyimbazi
J
Xu
X
et al
Interleukin-13: central mediator of allergic asthma.
Science.
282
1998
2258
2261
94
Grunig
G
Warnock
M
Wakil
AE
et al
Requirement for IL-13 independently of IL-4 in experimental asthma.
Science.
282
1998
2261
2263
95
Luttmann
W
Knoechel
B
Foerster
M
Matthys
H
Virchow
JC
Jr
Kroegel
C
Activation of human eosinophils by IL-13. Induction of CD69 surface antigen, its relationship to messenger RNA expression, and promotion of cellular viability.
J Immunol.
157
1996
1678
1683
96
Ying
S
Meng
Q
Barata
LT
Robinson
DS
Durham
SR
Kay
AB
Associations between IL-13 and IL-4 (mRNA and protein), vascular cell adhesion molecule-1 expression, and the infiltration of eosinophils, macrophages, and T cells in allergen-induced late-phase cutaneous reactions in atopic subjects.
J Immunol.
158
1997
5050
5057
97
Li
L
Xia
Y
Nguyen
A
et al
Effects of Th2 cytokines on chemokine expression in the lung: IL-13 potently induces eotaxin expression by airway epithelial cells.
J Immunol.
162
1999
2477
2487
98
Zhu
Z
Homer
RJ
Wang
Z
et al
Pulmonary expression of interleukin-13 causes inflammation, mucus hypersecretion, subepithelial fibrosis, physiologic abnormalities, and eotaxin production.
J Clin Invest.
103
1999
779
788
99
Dvorak
AM
Cell biology of the basophil.
Int Rev Cytol.
180
1998
87
236
100
Norman
JC
Price
LS
Ridley
AJ
Koffer
A
The small GTP-binding proteins, Rac and Rho, regulate cytoskeletal organization and exocytosis in mast cells by parallel pathways.
Mol Biol Cell.
7
1996
1429
1442
101
Kepley
CL
Wilson
BS
Oliver
JM
Identification of the FcepsilonRI-activated tyrosine kinases Lyn, Syk, and Zap-70 in human basophils.
J Allergy Clin Immunol.
102
1998
304
315
102
Kepley
CL
Youssef
L
Andrews
RP
Wilson
BS
Oliver
JM
Syk deficiency in nonreleaser basophils.
J Allergy Clin Immunol.
104
1999
279
284
103
Malveaux
FJ
Conroy
MC
Adkinson
NF
Jr
Lichtenstein
LM
IgE receptors on human basophils. Relationship to serum IgE concentration.
J Clin Invest.
62
1978
176
181
104
MacGlashan
D
Jr
McKenzie-White
J
Chichester
K
et al
In vitro regulation of FcepsilonRIalpha expression on human basophils by IgE antibody.
Blood.
91
1998
1633
1643
105
MacGlashan
DW
Jr
Bochner
BS
Adelman
DC
et al
Down-regulation of Fc(epsilon)RI expression on human basophils during in vivo treatment of atopic patients with anti-IgE antibody.
J Immunol.
158
1997
1438
1445
106
Knol
EF
Koenderman
L
Mul
E
Verhoeven
AJ
Roos
D
Differential mechanisms in the stimulus-secretion coupling in human basophils: evidence for a protein-kinase-C-dependent and a protein-kinase-C-independent route.
Agents Actions.
30
1990
49
52
107
Gibbs
BF
Grabbe
J
Inhibitors of PI 3-kinase and MEK kinase differentially affect mediator secretion from immunologically activated human basophils.
J Leukoc Biol.
65
1999
883
890
108
Ishizuka
T
Terada
N
Gerwins
P
et al
Mast cell tumor necrosis factor alpha production is regulated by MEK kinases.
Proc Natl Acad Sci U S A.
94
1997
6358
6363
109
Marquardt
DL
Alongi
JL
Walker
LL
The phosphatidylinositol 3-kinase inhibitor wortmannin blocks mast cell exocytosis but not IL-6 production.
J Immunol.
156
1996
1942
1945
110
Ishizuka
T
Oshiba
A
Sakata
N
Terada
N
Johnson
GL
Gelfand
EW
Aggregation of the FcepsilonRI on mast cells stimulates c-Jun amino-terminal kinase activity. A response inhibited by wortmannin.
J Biol Chem.
271
1996
12762
12766
111
Ozawa
K
Masujima
T
Ikeda
K
Kodama
Y
Nonomura
Y
Different pathways of inhibitory effects of wortmannin on exocytosis are revealed by video-enhanced light microscope.
Biochem Biophys Res Commun.
222
1996
243
248
112
Suzuki
M
Hirai
K
Kitani
S
et al
Pharmacologic study of basophil histamine release induced by monocyte chemotactic protein-1 with kinase inhibitors.
Int Arch Allergy Immunol.
111
1996
18
22
113
Schroeder
JT
Howard
BP
Jenkens
MK
Kagey-Sobotka
A
Lichtenstein
LM
MacGlashan
DW
Jr
IL-4 secretion and histamine release by human basophils are differentially regulated by protein kinase C activation.
J Leukoc Biol.
63
1998
692
698
114
Pendl
GG
Prieschl
EE
Thumb
W
Harrer
NE
Auer
M
Baumruker
T
Effects of phosphatidylinositol-3-kinase inhibitors on degranulation and gene induction in allergically triggered mouse mast cells.
Int Arch Allergy Immunol.
112
1997
392
399
115
Seger
R
Krebs
EG
The MAPK signaling cascade.
FASEB J.
9
1995
726
735
116
Miura
K
Schroeder
JT
Hubbard
WC
MacGlashan
DW
Jr
Extracellular signal-regulated kinases regulate leukotriene C4 generation, but not histamine release or IL-4 production from human basophils.
J Immunol.
162
1999
4198
4206
117
Schleimer
RP
Derse
CP
Friedman
B
et al
Regulation of human basophil mediator release by cytokines. I. Interaction with antiinflammatory steroids.
J Immunol.
143
1989
1310
1317
118
MacGlashan
DW
Jr
Hubbard
WC
IL-3 alters free arachidonic acid generation in C5a-stimulated human basophils.
J Immunol.
151
1993
6358
6369
119
Krieger
M
von Tscharner
V
Dahinden
CA
Signal transduction for interleukin-3-dependent leukotriene synthesis in normal human basophils: opposing role of tyrosine kinase and protein kinase.
Eur J Immunol.
22
1992
2907
2913
120
Miura
K
Hubbard
WC
MacGlashan
DW
Jr
Phosphorylation of cytosolic phospholipase A2 by IL-3 is associated with increased free arachidonic acid generation and leukotriene C4 release in human basophils.
J Allergy Clin Immunol.
102
1998
512
520
121
Lichtenstein
LM
Osler
AG
Studies on the mechanism of hypersensitivity phenomena: IX. Histamine release from human leukocytes by rageweed pollen antigen.
J Exp Med.
120
1964
507
530
122
Schroeder
JT
MacGlashan
DW
Jr
Kagey-Sobotka
A
White
JM
Lichtenstein
LM
IgE-dependent IL-4 secretion by human basophils. The relationship between cytokine production and histamine release in mixed leukocyte cultures.
J Immunol.
153
1994
1808
1817
123
MacGlashan
D
Jr
Desensitization of IgE-mediated IL-4 release from human basophils.
J Leukoc Biol.
63
1998
59
67
124
MacGlashan
D
Jr
Botana
LM
Biphasic Ca2+ responses in human basophils. Evidence that the initial transient elevation associated with the mobilization of intracellular calcium is an insufficient signal for degranulation.
J Immunol.
150
1993
980
991
125
Botana
LM
MacGlashan
DW
Differential effects of cAMP-elevating drugs on stimulus-induced cytosolic calcium changes in human basophils.
J Leukoc Biol.
55
1994
798
804
126
Schroeder
JT
MacGlashan Jr
DW
Lichtenstein
LM
Mechanisms and pharmacologic control of basophil-eerived IL-4 and IL-13.
Int Arch Allergy Immunol.
118
1999
87
89
127
Redrup
AC
Howard
BP
MacGlashan
DW
Jr
Kagey-Sobotka
A
Lichtenstein
LM
Schroeder
JT
Differential regulation of IL-4 and IL-13 secretion by human basophils: their relationship to histamine release in mixed leukocyte cultures.
J Immunol.
160
1998
1957
1964
128
Gibbs
BF
Vollrath
IB
Albrecht
C
Amon
U
Wolff
HH
Inhibition of interleukin-4 and interleukin-13 release from immunologically activated human basophils due to the actions of anti-allergic drugs.
Naunyn Schmiedebergs Arch Pharmacol.
357
1998
573
578
129
MacGlashan
DW
Jr
White
JM
Huang
S-K
Ono
SJ
Schroeder
JT
Lichtenstein
LM
Secretion of IL-4 from basophils. The relationship between IL-4 mRNA and protein in resting and stimulated basophils.
J Immunol.
152
1994
3006
3016
130
Kon
OM
Sihra
BS
Till
SJ
Corrigan
CJ
Kay
AB
Grant
JA
Unstimulated basophils in atopic and nonatopic subjects express intracellular interleukin-4: detection by flow cytometry.
Allergy.
53
1998
891
896
131
Geissler
EN
Ryan
MA
Housman
DE
The dominant-white spotting (W) locus of the mouse encodes the c-kit proto-oncogene.
Cell.
55
1988
185
192
132
Lantz
CS
Boesiger
J
Song
CH
et al
Role for interleukin-3 in mast-cell and basophil development and in immunity to parasites.
Nature.
392
1998
90
93
133
Abraham
SN
Arock
M
Mast cells and basophils in innate immunity.
Semin Immunol.
10
1998
373
381
134
Abraham
SN
Malaviya
R
Mast cells in infection and immunity.
Infect Immun.
65
1997
3501
3508
135
Patella
V
Giuliano
A
Bouvet
JP
Marone
G
Endogenous superallergen protein Fv induces IL-4 secretion from human Fc epsilon RI+ cells through interaction with the VH3 region of IgE.
J Immunol.
161
1998
5647
5655
136
Patella
V
Giuliano
A
Florio
G
Bouvet
JP
Marone
G
Endogenous superallergen protein Fv interacts with the VH3 region of IgE to induce cytokine secretion from human basophils.
Int Arch Allergy Immunol.
118
1999
197
199
137
Patella
V
Florio
G
Petraroli
A
Marone
G
HIV-1 gp120 induces IL-4 and IL-13 release from human Fc epsilon RI+ cells through interaction with the VH3 region of IgE.
J Immunol.
164
2000
589
595
138
Falcone
FH
Dahinden
CA
Gibbs
BF
et al
Human basophils release interleukin-4 after stimulation with Schistosoma mansoni egg antigen.
Eur J Immunol.
26
1996
1147
1155
139
Pruzansky
JJ
Grammer
LC
Pattterson
R
Roberts
M
Dissociation of IgE from receptors on human basophils. I. Enhanced passive sensitization for histamine release.
J.Immunol.
131
1983
1949
1953
140
Gregory
WF
Blaxter
ML
Maizels
RM
Differentially expressed, abundant trans-spliced cDNAs from larval Brugia malayi.
Mol Biochem Parasitol.
87
1997
85
95
141
MacDonald
SM
Rafnar
T
Langdon
J
Lichtenstein
LM
Molecular identification of an IgE-dependent histamine-releasing factor.
Science.
269
1995
688
690
142
Schroeder
JT
Lichtenstein
LM
MacDonald
SM
Recombinant histamine-releasing factor enhances IgE-dependent IL-4 and IL-13 secretion by human basophils.
J Immunol.
159
1997
447
452
143
MacDonald
SM
Human recombinant histamine-releasing factor.
Int Arch Allergy Immunol.
113
1997
187
189
144
Wantke
F
MacGlashan
DW
Langdon
JM
MacDonald
SM
The human recombinant histamine releasing factor: functional evidence that it does not bind to the IgE molecule.
J Allergy Clin Immunol.
103
1999
642
648
145
Haas
H
Falcone
FH
Holland
MJ
et al
Early interleukin-4: its role in the switch towards a Th2 response and IgE-mediated allergy.
Int Arch Allergy Immunol.
119
1999
86
94
146
Coffman
RL
von der Weid
T
Multiple pathways for the initiation of T helper 2 (Th2) responses.
J Exp Med.
185
1997
373
375
147
Siraganian
RP
Siragania
PA
Mechanism of action of concanavalin A on human basophils.
J Immunol.
114
1975
886
893
148
Shibasaki
M
Sumazaki
R
Isoyama
S
Takita
H
Interaction of lectins with human IgE: IgE-binding property and histamine-releasing activity of twelve plant lectins.
Int Arch Allergy Immunol.
98
1992
18
25
149
Haas
H
Falcone
FH
Schramm
G
et al
Dietary lectins can induce in vitro release of IL-4 and IL-13 from human basophils.
Eur J Immunol.
29
1999
918
927
150
Loukas
A
Maizels
RM
Helminth C-type lectins and host-parasite interactions.
Parasitol Today.
16
2000
333
339
151
Loukas
A
Mullin
NP
Tetteh
KK
Moens
L
Maizels
RM
A novel C-type lectin secreted by a tissue-dwelling parasitic nematode.
Curr Biol.
12
1999
825
828
152
Baenziger
J
Kornfeld
S
Structure of the carbohydrate units of IgE immunoglobulin. I. Over-all composition, glycopeptide isolation, and structure of the high mannose oligosaccharide unit.
J Biol Chem.
249
1974
1889
1896
153
Baenziger
J
Kornfeld
S
Structure of the carbohydrate units of IgE immunoglobulin. II. Sequence of the sialic acid -containing glycopeptides.
J Biol Chem.
249
1974
1897
1903
154
Pritchard
DI
Brown
A
Kasper
G
et al
A hookworm allergen which strongly resembles calreticulin.
Parasite Immunol.
21
1999
439
450
155
Krause
KH
Michalak
M
Calreticulin.
Cell.
88
1997
439
443
156
Koshino
T
Arai
Y
Miyamoto
Y
et al
Airway basophil and mast cell density in patients with bronchial asthma: relationship to bronchial hyperresponsiveness.
J Asthma.
33
1996
89
95
157
Castells
MC
Irani
AM
Schwartz
LB
Evaluation of human peripheral blood leukocytes for mast cell tryptase.
J Immunol.
138
1987
2184
2189
158
Bieber
T
de la Salle
H
Wollenberg
A
et al
Human epidermal Langerhans cells express the high affinity receptor for immunoglobulin E (Fc epsilon RI).
J Exp Med.
175
1992
1285
1290
159
Maurer
D
Fiebiger
S
Ebner
C
et al
Peripheral blood dendritic cells express Fc epsilon RI as a complex composed of Fc epsilon RI alpha- and Fc epsilon RI gamma-chains and can use this receptor for IgE-mediated allergen presentation.
J Immunol.
157
1996
607
616
160
Gounni
AS
Lamkhioued
B
Ochiai
K
et al
High affinity IgE receptor on eosinophils is involved in defence against parasites.
Nature.
367
1994
183
186
161
Maurer
D
Fiebiger
E
Reininger
B
et al
Expression of functional high affinity immunoglobulin E receptors (Fc epsilon RI) on monocytes of atopic individuals.
J Exp Med.
179
1994
745
750
162
Xia
HZ
Kepley
CL
Sakai
K
Chelliah
J
Irani
AM
Schwartz
LB
Quantitation of tryptase, chymase, Fc epsilon RI alpha, and Fc epsilon RI gamma mRNAs in human mast cells and basophils by competitive reverse transcription-polymerase chain reaction.
J Immunol.
154
1995
5472
5480
163
Bodger
MP
Mounsey
GL
Nelson
J
Fitzgerald
PH
A monoclonal antibody reacting with human basophils.
Blood.
69
1987
1414
1418
164
Kepley
CL
Craig
SS
Schwartz
LB
Identification and partial characterization of a unique marker for human basophils.
J Immunol.
154
1995
6548
6555
165
Irani
AM
Huang
C
Xia
HZ
et al
Immunohistochemical detection of human basophils in late-phase skin reactions.
J Allergy Clin Immunol.
101
1998
354
362
166
Hedrick
JL
Smith
AJ
Size and charge isomer separation and estimation of molecular weights of proteins by disc gel electrophoresis.
Arch Biochem Biophys.
126
1968
155
164
167
McEuen
AR
Buckley
MG
Compton
SJ
Walls
AF
Development and characterization of a monoclonal antibody specific for human basophils and the identification of a unique secretory product of basophil activation.
Lab Invest.
79
1999
27
38
168
Kishi
K
A new leukemia cell line with Philadelphia chromosome characterized as basophil precursors.
Leuk Res.
9
1985
381
390
169
Blom
T
Huang
R
Aveskogh
M
Nilsson
K
Hellman
L
Phenotypic characterization of KU812, a cell line identified as an immature human basophilic leukocyte.
Eur J Immunol.
22
1992
2025
2032
170
Nilsson
G
Carlsson
M
Jones
I
Ahlstedt
S
Matsson
P
Nilsson
K
TNF-alpha and IL-6 induce differentiation in the human basophilic leukaemia cell line KU812.
Immunology.
81
1994
73
78
171
Navarro
S
Louache
F
Debili
N
Vainchenker
W
Doly
J
Autocrine regulation of terminal differentiation by interleukin-6 in the pluripotent KU812 cell line.
Biochem Biophys Res Commun.
169
1990
184
191
172
Valent
P
Besemer
J
Kishi
K
et al
IL-3 promotes basophilic differentiation of KU812 cells through high affinity binding sites.
J Immunol.
145
1990
1885
1889
173
Hara
T
Yamada
K
Tachibana
H
Basophilic differentiation of the human leukemia cell line KU812 upon treatment with interleukin-4.
Biochem Biophys Res Commun.
247
1998
542
548
174
Seigneurin
D
Champelovier
P
Mouchiroud
G
et al
Human chronic myeloid leukemic cell line with positive Philadelphia chromosome exhibits megakaryocytic and erythroid characteristics.
Exp Hematol.
15
1987
822
832
175
Blom
T
Nilsson
G
Sundström
C
Nilsson
K
Hellman
L
Characterization of a human basophil-like cell line (LAMA-84).
Scand J Immunol.
44
1996
54
61
176
Kepley
CL
Pfeiffer
JR
Schwartz
LB
Wilson
BS
Oliver
JM
The identification and characterization of umbilical cord blood-derived human basophils.
J Leukoc Biol.
64
1998
474
483
177
Haisch
K
Gibbs
BF
Korber
H
et al
Purification of morphologically and functionally intact human basophils to near homogeneity.
J Immunol Methods.
226
1999
129
137
178
Gibbs
BF
Noll
T
Falcone
FH
et al
A three-step procedure for the purification of human basophils from buffy coat blood.
Inflamm Res.
46
1997
137
142
179
Schroeder
JT
Hanrahan
LRJ
Purification of human basophils using mouse monoclonal IgE.
J Immunol Methods.
133
1990
269
277
180
Weil
GJ
Leiserson
WM
Chused
TM
Isolation of human basophils by flow microfluorometry.
J Immunol Methods.
58
1983
359
363
181
Mul
FP
Knol
EF
Roos
D
An improved method for the purification of basophilic granulocytes from human blood.
J Immunol Methods.
149
1992
207
214
182
Bjerke
T
Nielsen
S
Helgestad
J
Nielsen
BW
Schiotz
PO
Purification of human blood basophils by negative selection using immunomagnetic beads.
J Immunol Methods.
157
1993
49
56
183
Tsang
S
Hayashi
M
Zheng
X
Campbell
A
Schellenberg
RR
Simplified purification of human basophils.
J Immunol Methods.
233
2000
13
20
184
Ponath
PD
Qin
S
Ringler
DJ
et al
Cloning of the human eosinophil chemoattractant, eotaxin. Expression, receptor binding, and functional properties suggest a mechanism for the selective recruitment of eosinophils.
J Clin Invest.
97
1996
604
612
185
Hogaboam
C
Kunkel
SL
Strieter
RM
et al
Novel role of transmembrane SCF for mast cell activation and eotaxin production in mast cell-fibroblast interactions.
J Immunol.
160
1998
6166
6171
186
Gao
JL
Sen
AI
Kitaura
M
et al
Identification of a mouse eosinophil receptor for the CC chemokine eotaxin.
Biochem Biophys Res Commun.
223
1996
679
684
187
Kitaura
M
Nakajima
T
Imai
T
et al
Molecular cloning of human eotaxin, an eosinophil-selective CC chemokine, and identification of a specific eosinophil eotaxin receptor, CC chemokine receptor 3.
J Biol Chem.
271
1996
7725
7730
188
Sallusto
F
Mackay
CR
Lanzavecchia
A
Selective expression of the eotaxin receptor CCR3 by human T helper 2 cells.
Science.
277
1997
2005
2007
189
Robinson
DS
Hamid
Q
Ying
S
et al
Predominant TH2-like bronchoalveolar T-lymphocyte population in atopic asthma.
N Engl J Med.
326
1992
298
304
190
Ochi
H
Hirani
WM
Yuan
Q
Friend
DS
Austen
KF
Boyce
JA
T helper cell type 2 cytokine-mediated comitogenic responses and CCR3 expression during differentiation of human mast cells in vitro.
J Exp Med.
190
1999
267
280
191
Romagnani
P
De Paulis
A
Beltrame
C
et al
Tryptase-chymase double-positive human mast cells express the eotaxin receptor CCR3 and are attracted by CCR3-binding chemokines.
Am J Pathol.
155
1999
1195
1204

Author notes

Bernhard F. Gibbs, Department of Dermatology, Medical University of Lübeck, Ratzeburger Allee 160, D-23538 Lübeck, Germany.

Sign in via your Institution