We investigated the role of Bruton's tyrosine kinase (Btk) in FcεRI-dependent activation of mouse mast cells, using xid and btk null mutant mice. Unlike B cell development, mast cell development is apparently normal in these btk mutant mice. However, mast cells derived from these mice exhibited significant abnormalities in FcεRI-dependent function. xid mice primed with anti-dinitrophenyl monoclonal IgE antibody exhibited mildly diminished early-phase and severely blunted late-phase anaphylactic reactions in response to antigen challenge in vivo. Consistent with this finding, cultured mast cells derived from the bone marrow cells of xid or btk null mice exhibited mild impairments in degranulation, and more profound defects in the production of several cytokines, upon FcεRI cross-linking. Moreover, the transcriptional activities of these cytokine genes were severely reduced in FcεRI-stimulated btk mutant mast cells. The specificity of these effects of btk mutations was confirmed by the improvement in the ability of btk mutant mast cells to degranulate and to secrete cytokines after the retroviral transfer of wild-type btk cDNA, but not of vector or kinase-dead btk cDNA. Retroviral transfer of Emt (= Itk/Tsk), Btk's closest relative, also partially improved the ability of btk mutant mast cells to secrete mediators. Taken together, these results demonstrate an important role for Btk in the full expression of FcεRI signal transduction in mast cells.

Mast cells and basophils play pivotal roles in the initiation of allergic reactions. Cross-linking of the high-affinity receptor for IgE (FcεRI) on these cells activates intracellular signaling pathways that lead to degranulation and release of histamine and other preformed mediators, de novo synthesis and release of lipid mediators, and secretion of preformed and de novo synthesized cytokines (1, 2). These bioactive mediators are thought to lead to allergic inflammation.

FcεRI consists of one molecule of an α subunit that is capable of binding to IgE, one molecule of a β subunit with four transmembrane segments, and two molecules of disulfide-bonded γ subunits (3). None of these subunits have discernible enzyme structures, but both the β and γ subunits have the immunoreceptor tyrosine-based activation motif (ITAM; references 4, 5).1 After FcεRI cross-linking, tyrosine phosphorylation of several intracellular proteins is the earliest recognizable activation event (6). The importance of protein tyrosine kinases (PTKs) in FcεRI-mediated mediator secretion has been demonstrated by showing that treatment with a variety of PTK inhibitors can abrogate FcεRI-dependent activation of mast cells (7, 8). Two specific PTKs, Lyn and Syk, that belong to the Src and Syk/ZAP families, respectively, were shown to be essential for FcεRI-mediated mast cell activation (911). According to a generally accepted hypothesis (12), Lyn that is associated with the β subunit in unstimulated cells is activated upon FcεRI cross-linking. Subsequently, activated Lyn phosphorylates tyrosine residues within the ITAM sequences in the β and γ subunits. Phosphorylated ITAM (phospho-ITAM) in the β subunit recruits new molecules of Lyn through the Src homology 2 (SH2) domain–phosphotyrosine interaction while phospho-ITAM in the γ subunit recruits Syk by the same mechanism (13). Lyn and Syk are activated when bound to phospho-ITAMs (14, 15), and such activated Lyn and Syk in turn phosphorylate downstream targets such as phospholipase C (PLC)–γ.

Three Tec family PTKs, Btk, Emt/Itk/Tsk (Emt), and Tec, are also expressed in mast cells (16, 17). Among them, Btk and Emt are activated upon FcεRI cross-linking, suggesting a functional role in mast cell activation (18, 19). However, in contrast with Lyn and Syk (2022), these PTKs do not appear to be receptor-associated molecules. Moreover, both Btk and Emt have important roles that are apparently unrelated to their involvement in FcεRI-dependent mast cell activation. Thus, Btk plays an essential role in the differentiation and activation of B lymphocytes: defects in the btk gene lead to X-linked agammaglobulinemia in humans (23, 24) and X-linked immunodeficiency (xid) in mice (25, 26). In addition, subsequent studies have implicated Btk in a number of signal transduction pathways in immune cells, including those for the B cell antigen receptor (2729), CD38 (30, 31), CD40 (32), IL-5 (33), IL-6 (34), and IL-10 (35). Emt is considered a “T cell equivalent” of Btk, and is involved in T cell development and early activation events triggered through TCR/CD3 and CD28 (3638).

Both xid (a mutation which results in the substitution of Arg with Cys at residue 28 in the Btk protein) and btk null mice exhibit essentially the same phenotype: these mutations lead to reduced numbers of mature conventional B cells, a severe deficiency of B1 B cells, a deficiency of serum IgM and IgG3, and defective responses to various B cell activators in vitro and to immunization with thymus-independent type II antigens in vivo (39, 40).

In this study, we analyzed Btk functions in mast cells in vivo and in vitro. Although btk mutant mast cells appear normal in many aspects of development in vitro or in vivo, they exhibited multiple abnormalities in FcεRI-mediated functions. Btk mutant mast cells exhibited mild to moderate impairment of FcεRI-mediated degranulation and histamine release, and more severe impairment of FcεRI-mediated cytokine production in vitro. Btk mutant mice exhibited correspondingly mild versus severe abnormalities in the early versus late phases of FcεRI-mediated cutaneous inflammatory responses in vivo. Furthermore, we found that both xid and null mutations of the btk gene result in defects in the transcriptional regulation of cytokine genes in mast cells stimulated via FcεRI, and such defects in btk mutant mast cells could be improved by retroviral gene transfer of wild-type (wt) btk cDNA. These results collectively demonstrate the involvement of Btk in the full expression of FcεRI signal transduction.

Passive Cutaneous Anaphylactic (PCA) Reactions.

In homologous PCA experiments, 10 μl of various amounts of anti-DNP monoclonal IgE was intradermally injected into the ear of mice. 24 h later, 0.25 ml saline solution containing 1 mg/ml DNP conjugates of BSA (DNP8.7-BSA) and 0.5% Evans blue dye was intravenously injected. The amounts of extravasated dye were measured after 30 min by extracting ears with potassium hydroxide as previously described (41). In another type of experiment, CBA/J and CBA/CaHN-xid/J mice received 1.0 ml anti-DNP monoclonal IgE antibody intravenously. 24 h later, a skin reaction was elicited by applying 10 μl 0.75% dinitrofluorobenzene acetone– olive oil solution to both sides of the ears. The reaction was assessed by measuring the ear thickness using an engineer's micrometer, Upright Dial Gauge (Peacock, Tokyo, Japan), at the indicated times after antigen challenge (42).

Cell Culture and Stimulation.

Bone marrow cells taken from mouse femurs were incubated in the presence of IL-3 as previously described (7). After 4 wk of culture, cells (>95% mast cells, termed BMMCs for bone marrow–derived cultured mast cells) were incubated overnight with anti-DNP IgE antibody. Unless otherwise indicated (e.g., in cells stimulated for release of histamine or leukotrienes, see below), sensitized cells were stimulated for 24 h with 30 ng/ml DNP conjugates of human serum albumin (DNP-HSA) in RPMI 1640 medium supplemented with 10% fetal bovine serum, 50 μM 2-ME, 2 mM glutamine, and IL-3. For most retroviral transfection experiments, bone marrow cells cultured in the presence of IL-3 for 2–3 wk were expanded in the presence of both IL-3 and recombinant rat stem cell factor (SCF; gift of Kirin Brewery Co., Tokyo, Japan) for another 1–2 wk. At this point, >95% of the cells were mast cells, termed sBMMCs (for SCF-maintained BMMCs).

Northern Blot Analysis.

Total cellular RNAs were isolated using RNAzol B (Tel-Test, Inc., Friendswood, TX) according to the manufacturer's instructions. RNAs fractionated by formaldehyde/agarose gel electrophoresis were blotted onto nitrocellulose membranes. Mouse TNF-α (obtained from the American Type Culture Collection, Rockville, MD) and c-myc (a gift from D.R. Green, La Jolla Institute for Allergy and Immunology, La Jolla, CA) cDNA fragments were gel-purified and 32P-labeled with a Megaprime DNA labeling kit (Pharmacia Biotech, Piscataway, NJ). Membranes were hybridized with 32P-labeled probe purified through Elutip-d (Schleicher & Schuell, Keene, NH). Hybridized bands were detected by autoradiography.

Immunoblot Analysis.

Immunologically stimulated cells were lysed in 1% NP-40–containing buffer. Cleared lysates were directly analyzed by SDS-PAGE or immunoprecipitated with polyclonal anti–TNF-α antibodies (Genzyme Corp., Cambridge, MA) before SDS-PAGE. Proteins in gels were electrophoretically transferred to Immobilon-P membranes (Millipore Corp., Bedford, MA). Membranes were blocked, incubated with anti–TNF-α, anti-Btk (43), anti-Emt (44), or other appropriate primary antibodies, and then with horseradish peroxidase–conjugated secondary antibody. Immunoreactive bands were detected by an enhanced chemiluminescence kit (Amersham Corp., Arlington Heights, IL).

Transfection.

Murine btk cDNA in pME18S vector (16) was used for in vitro mutagenesis using two-step PCR procedures (45) to generate K430R and other btk mutants. The wt and mutant btk cDNAs confirmed by sequencing were inserted into the Moloney murine leukemia virus–based retroviral vectors, pMX-neo or pMX-puro (46). Retroviruses were generated by transient transfection of BOSC-23 packaging cells (47) with Lipofectamine (GIBCO BRL, Gaithersburg, MD). BMMCs or sBMMCs derived from male xid or btk null mice were infected with these retroviruses in the presence of 10 μg/ml polybrene. Selection with G418 (for xid-BMMCs and xid-sBMMCs) or puromycin (for btk null–BMMCs and btk null–sBMMCs) was started 48 h after infection. Mass populations of G418- or puromycin-resistant cells were grown and then cultured in the absence of selection drug for 48 h before immunological stimulation.

Measurements of Secreted Histamine, Cytokines, and Leukotrienes.

Histamine released into the media during a 45-min stimulation was measured by an automatic fluorometric assay (48). Concentrations of antigen (ED50) for half maximal histamine release was estimated using 77% (wt) and 79% (xid) as maximal responses. TNF-α, IL-2, IL-4, IL-6, and GM-CSF secreted into the media for 24 h were measured by ELISA kits (Endogen, Woburn, MA). Leukotrienes secreted into media for 30 min were analyzed by an enzyme immunoassay kit for leukotriene C4/D4/E4 (Amersham Corp.).

Transcriptional Activity Assay with Luciferase Reporter Constructs.

Luciferase reporter constructs, mouse IL-2 (−321), nuclear factor of activated T cells (NFAT)d–luc, NFκB–luc, and c-fos–luc have been previously described (49, 50). To engineer the human TNF-α (−200)–luc, PCR was done to amplify a DNA fragment containing the TNF-α promoter region (−199 to +68). This PCR fragment was inserted into the SmaI/BglII site of pGL3-Basic vector (Promega Corp., Madison, WI). 1.0–1.5 × 107 mast cells were transfected with 5–10 μg reporter plasmids by electroporation at 400 V, 950 μF using a Gene Pulser II apparatus (Bio-Rad, Hercules, CA). Transfected cells were sensitized with anti-DNP monoclonal IgE antibody overnight, and left unstimulated or stimulated with 30 ng/ml DNP-HSA for 8 h before cell harvest. Cells were lysed in 0.2% Triton X-100 in 100 mM potassium phosphate buffer (pH 7.8)/1 mM dithiothreitol. Luminescence of cleared lysates was measured after addition of luciferin solution using a luminometer (Monolight 2010; Analytical Luminescence Laboratory, San Diego, CA).

Quantitation of Tissue Mast Cells.

Tissue mast cells in ear skin were quantified by light microscopy at ×400 by an observer who was unaware of the identity (i.e., mouse genotype) of the individual specimens, in 1-μm, Epon-embedded, Giemsa-stained sections, as previously described (51). Results were expressed as mast cells (mean ± SEM) per mm2 of dermis (51).

Mast Cell Development Is Not Affected by btk Mutations.

First, we assessed the effects of btk null and xid mutations on several aspects of mast cell development and phenotype in vivo or in vitro. Mast cells in wt and btk null mouse ear skins were similar in their morphology and anatomical distribution (data not shown) and numbers: 125 ± 27.9/mm2 of dermis (129/C57BL F2) versus 123 ± 32.2/mm2 of dermis (btk null). The phenotypes of BMMCs were indistinguishable between wt (CBA/J) and xid (CBA/HCaN-xid/J) as well as between wt (129/C57BL F2) and btk null mice in their morphology when the cells were stained with May-Giemsa or with Alcian Blue (data not shown), and in numbers of IgE binding sites: (4.6 ± 1.2) × 104/cell (CBA/J) versus (3.7 ± 2.0) × 104/cell (CBA/HCaN-xid/J); (7.9 ± 2.4) × 104/cell (129/C57BL F2) versus (8.2 ± 2.9) × 104/ cell (btk null). The wt- and btk null–BMMCs were similar in the expression of various signaling proteins, including FcεRI β and γ subunits, Lyn, Syk, Grb2, Shc, Sos, H-Ras, PLC-γ1, SPY75 (= HS1), protein kinase C (PKC; α, βI, βII, δ, ε, η, θ, and ζ isoforms), ERK1/2, JNK1/2, p38, PAK65, SEK1, and c-Jun (data not shown). Therefore, we concluded that either btk null or xid mutations apparently do not significantly interfere multiple aspects of mast cell development in vivo and in vitro.

Effects of btk Mutations on Anaphylactic Reactions In Vivo.

We next tested the effects of the btk mutations on mast cell activation events induced by FcεRI cross-linking. Two types of PCA experiments were carried out. Mice primed by intradermal injection of anti-DNP IgE for 24 h were injected intravenously with antigen and Evans blue dye. Extravasation of Evans blue dye, due to increased blood vessel permeability as a result of PCA reactions, was quantified. The extravasation of Evans blue dye during the first 30 min of the PCA reactions, which is dependent mainly on histamine and serotonin released from activated mast cells (52), was slightly but significantly reduced at all the tested IgE doses in xid mice compared with wt mice (Fig. 1,A). To examine another type of PCA reaction (42), mice were sensitized with anti-DNP IgE 24 h before a solution of 0.75% dinitrofluorobenzene (hapten) was applied epicutaneously to the ear skin. xid mice exhibited little or no IgE/ antigen-specific edema, whereas wt mice exhibited a prominent response that was detectable 4 h or later after antigen stimulation (Fig. 1,B). This late reaction is known to be at least partly due to TNF-α secreted from activated mast cells (51). Indeed, injection into the PCA-inducing site of wt mice with a neutralizing antibody to TNF-α just before antigen application significantly suppressed the development of the edema associated with the late phase of the reaction, as measured 24 h after antigen stimulation (Fig. 1 C). Significant defects in both the early and late phases of PCA reactions were also observed in btk null mice (data not shown).

Effects of btk Mutations on FcεRI-mediated Degranulation and Cytokine Secretion.

To investigate the cellular basis for the diminished PCA reactions in btk mutant mice, the capacities to degranulate and release histamine and to produce and secrete cytokines were compared between BMMCs derived from the wt mice and the xid (or btk null) mice. Consistent with the modest defect in the early phase of PCA reactions and the more striking defect in the later phase, xid-BMMCs showed relatively mild defects in FcεRI-elicited histamine release but more severe impairments in cytokine secretion compared with wt-BMMCs. Maximal histamine responses (70–80% of the cellular content) were similar between wt- and xid-BMMCs. However, xid-BMMCs exhibited a marked reduction in histamine release at suboptimal doses of antigen, and the sensitivity of xid-BMMCs to antigen stimulation was reduced by 3.8-fold compared with wt-BMMCs (ED50 = 4.4 ng/ml [wt] versus 17 ng/ml [xid], see Fig. 2,A). Btk null–BMMCs exhibited a somewhat more severe defect with a reduction in maximal histamine release in addition to reduced antigen sensitivity (Fig. 2 B).

In contrast to the relatively mild defect in histamine release, the differences in TNF-α secretion between the xid- and wt-BMMCs stimulated with an optimal concentration (30 ng/ml) of antigen ranged between 1:3.2 and 1:12 (a mean of 1:6.7, n = 5, see Fig. 2,C). FcεRI-stimulated secretion of IL-2, IL-6, and GM-CSF was also impaired to a similar extent in xid-BMMCs (Fig. 2 C and data not shown). Similarly reduced cytokine responses were observed in btk null cells (data not shown). Wt-, xid-, and btk null–BMMCs secreted barely detectable amounts (<40 pg/ml) of IL-4 in response to an immunologic stimulation through FcεRI (data not shown). These results suggest that the abnormalities in the expression of PCA reactions in xid and btk null mice reflect the mildly reduced degranulation and markedly defective cytokine secretion exhibited by btk mutant mast cells upon FcεRI cross-linking.

Defects in the Transcription of Cytokine Genes in btk Mutant Mast Cells.

To further characterize the defects in TNF-α production and secretion in xid-BMMCs, we analyzed levels of TNF-α mRNA and protein in xid- and corresponding wt-BMMCs. As revealed by Northern blotting (Fig. 3 A), in wt-BMMCs, TNF-α mRNA was almost undetectable before stimulation but increased dramatically within 1 h after FcεRI cross-linking and decreased within the next few hours. However, under the same conditions of stimulation, xid-BMMCs produced less than one fifth the amount of TNF-α mRNA at its peak.

The membrane-bound TNF-α precursor (53), which was barely detectable before stimulation, increased after FcεRI cross-linking and reached a plateau level by 2–3 h after stimulation in wt-BMMCs (Fig. 3 B and data not shown). However, stimulation of xid-BMMCs led to only a slight increase in membrane-bound TNF-α content. Cellular pulse and chase experiments with [35S]methionine showed that there was no significant difference in the intracellular stability of TNF-α protein between wt- and xid-BMMCs (data not shown).

Taken together, these data suggest that Btk regulates TNF-α production at the transcriptional level. To test this possibility directly, we transfected BMMCs with luciferase reporter constructs under the control of TNF-α or IL-2 promoters. Transcription of both the TNF-α and IL-2 reporter constructs was strongly induced when wt-BMMCs were stimulated by FcεRI cross-linking. In btk null–BMMCs, the induced transcriptional activity of the TNF-α (−200)– luc construct was ∼50% of that in wt-BMMCs (Fig. 4,A). Induction of transcriptional activities of the IL-2 (−321)– luc construct was 4–5-fold less in btk null–BMMCs compared with its activity in wt cells (Fig. 4,A). Similar results were obtained when the transcriptional activity of the TNF-α and IL-2 constructs was assessed in xid- versus wt-BMMCs (data not shown). We then performed additional experiments to assess the specificity of the transcriptional regulation of cytokine genes by Btk. We found that btk null–sBMMCs that had been stably transfected with wt btk exhibited higher transcriptional activities of the IL-2 (−321)–luc, TNF-α (−200)–luc, and NFAT–luc constructs than the cells transfected with vector or kinase-dead (K430R) btk (Fig. 4,B). By contrast, all three cell populations exhibited similar low levels of activity for the NFκB and c-fos constructs (Fig. 4 B).

The NFAT family of transcription factors and AP-1 proteins play essential roles in the expression of the IL-2 (54, 55) and TNF-α genes (5658). These results indicate that defects in the production/secretion of cytokines upon FcεRI cross-linking in btk mutant mast cells are due, at least in part, to the inefficient transcription of these genes and may involve the signal transduction pathways leading to the activation of NFAT and/or AP-1 (Jun–Fos or Jun– Jun dimers). This notion is consistent with our recent data that Btk regulates JNK, an activator of c-Jun (59).

Gene Transfer–mediated Enhancement of the Ability of btk Mutant Mast Cells to Secrete Cytokines and Degranulate.

To further investigate the relationship between btk mutations and impairment of mast cell functions, we measured cytokine production in btk mutant mast cells that had been reconstituted with Btk by stable or transient transfection. For most of these experiments, we used mast cells (sBMMCs) that had been expanded in the presence of both IL-3 and SCF. When xid-sBMMCs that had been transfected with wt btk cDNA were stimulated by FcεRI cross-linking, we observed a substantial enhancement of TNF-α–producing/ secretory ability as compared to that seen in xid-sBMMCs that had been transfected with neo vector alone, with xid or kinase-dead (K430R) mutant btk cDNAs, or with wt syk or wt lyn cDNAs (Fig. 5,A). Expression of the transfected genes at comparable levels was confirmed by increased immunoreactive Btk proteins in wt, xid, and K430R mutant btk–transfected cells (Fig. 5,D, left). Transfectants expressing the constitutively active Btk* protein with the E41K substitution (60) exhibited somewhat higher levels of TNF-α secretion than did wt btk transfectants (Fig. 5 A). Moreover, none of the nontransfected or transfected sBMMCs secreted TNF-α without FcεRI stimulation (data not shown). Results similar to those shown for TNF-α secretion were also obtained when we tested the effects of transfection with wt versus various mutant btk cDNAs on the ability of FcεRI-stimulated xid-sBMMCs to secrete IL-2, IL-6, and GM-CSF (data not shown).

We next investigated btk null–sBMMCs transfectants, and in particular analyzed the domain requirements for Btk function in FcεRI-mediated cytokine production as opposed to degranulation. As shown in Fig. 5,B, transfection with wt btk cDNA greatly enhanced the ability of the cells to produce TNF-α, IL-2, IL-6, or GM-CSF in response to FcεRI-dependent activation, whereas compared with transfection with vector, transfection with kinase-dead (K430R) or SH2 mutant (R307K) btk cDNA had little or no effect. On the other hand, relatively low levels of protein expression for the product of the SH2 mutant (R307K) were detected in the stably transfected cells (Fig. 5 D, middle), indicating that the potential effects of this mutant btk in this system have not yet been adequately tested. Notably, transient transfection of btk null–sBMMCs with wt, but not kinase-dead, btk also restored cytokine gene transcriptional activities (data not shown). This finding provides further support for the conclusion that the results reflect the role of Btk in FcεRI signaling, not any role it might have in mast cell differentiation.

Notably, two mutant Btk proteins appeared to be able to partially (for TNF-α, IL-2 and GM-CSF) or fully (for IL-6) normalize 24-h cytokine production with respect to that seen in cells that had been transfected with wt btk cDNA (Fig. 5 B). One of these, the P265L mutation in the SH3 domain, is equivalent to the function-negative mutation in the SH3 domain of sem-5 (61). Therefore, this result suggests that at least partial FcεRI-dependent cytokine secretory function can be expressed in the absence of normal Btk SH3 function.

The other mutant btk cDNA that partially restored cytokine secretory ability in btk null–sBMMCs was xid (Fig. 5 B). This result may be related to the fact that levels of xid Btk protein in btk null–sBMMCs that had been transfected with xid btk cDNA were ∼20–30% greater than levels of wt Btk protein in the corresponding wt btk transfectants (by contrast, xid-BMMCs express only 1/5 to 1/3 the amount of Btk protein as do wt-BMMCs, data not shown). Thus, in comparison to xid-BMMCs, btk null–sBMMCs that had been transfected with xid btk cDNA greatly overexpress the xid Btk.

We previously noted (in Fig. 2) that the defect in FcεRI-dependent degranulation and histamine release exhibited by xid-BMMCs was less severe than that observed with btk null–BMMCs. Indeed, at optimal levels of FcεRI-dependent stimulation, xid-BMMCs gave a histamine release response that was indistinguishable from that of the corresponding wt-BMMCs (Fig. 2,A). We therefore examined the effect of transfection of btk null–sBMMCs with xid btk and other mutant btks, as opposed to wt btk, on degranulation (as assessed by histamine release) at optimal conditions of IgE sensitization and antigen challenge (Fig. 5,C). We found that the profound defect in the histamine release response of btk null–sBMMCs under these conditions was nearly fully restored by wt or xid btk cDNAs, was slightly enhanced by SH2 (R307K) or SH3 (P265L) mutant btk cDNAs, but was unaffected (relative to results obtained with vector alone) by the kinase-dead (K430R) mutant btk cDNA (Fig. 5 C).

Since Emt is not only closely related to Btk but is activated upon FcεRI cross-linking (19), we also examined whether Emt might influence defects in cytokine production or degranulation in btk null–sBMMCs. We found that btk null–sBMMCs express endogenous Emt protein (Fig. 5,D). Moreover, the overexpression of wt Emt protein, but not kinase-dead (K390R) Emt protein, enhanced both FcεRI-dependent cytokine production (Fig. 5,B) and histamine release (Fig. 5,C) in btk null–sBMMCs. However, transfection of btk null–sBMMCs with wt emt did not restore either cytokine production or histamine release to levels observed in cells which had been transfected with wt btk (Fig. 5, B and C).

Btk Functions in Mast Cells and Other Hematopoietic Cells.

Btk has been shown to have essential roles in B cell differentiation and activation. Although our in vivo and in vitro studies have thus far revealed no significant effects of the btk mutations on mast cell development, we have identified multiple defects in FcεRI-induced activation events in btk mutant mast cells. Both degranulation, leading to release of histamine, and production/secretion of several cytokines were mildly or severely impaired, respectively. These defects at the cellular levels probably account for the defective expression of anaphylactic reactions in response to IgE and antigen in btk mutant mice. Together with our previous data demonstrating the tyrosine phosphorylation and enzymatic activation of Btk upon FcεRI cross-linking (18), these in vivo and in vitro effects of btk mutations have established that Btk has a role in the expression of FcεRI-dependent mast cell function.

Notably, some of the effects of btk mutations are milder in mice than in humans. For example, X-linked agammaglobulinemia patients have few mature B cells with no or little immunoglobulin production, whereas xid or btk null mice have about half the number of B cells as in normal mice (39, 40, 62, 63). Such species differences in the consequences of btk mutations raise the possibility that the effects of btk mutations on mast cell development or function might be milder in mice than in humans. This possibility is currently being investigated.

The btk gene is also expressed in myeloid cells in addition to mast and B cells (16). Hence, we examined the ability of activated macrophages from btk mutant mice to secrete TNF-α. We found that lipopolysaccharide stimulation induced the secretion of indistinguishably high levels of TNF-α from wt versus xid mouse bone marrow–derived macrophages (98% Mac-1+) cultured in GM-CSF (data not shown). Therefore, the production/secretion of TNF-α seems to be differentially regulated in the two types of cells; it is more dependent on Btk in mast cells than in macrophages. This is not surprising, given the recent data demonstrating that the transcription of the TNF-α gene is regulated in a cell type–specific manner in activated T and B cells (58).

Structural Requirements of Btk for FcεRI-dependent Degranulation and Cytokine Production/Secretion.

Btk and Emt have, in order from their NH2 to COOH termini, pleckstrin homology (PH), Tec homology (TH), SH3, SH2, and SH1 (= kinase) domains. The catalytic activity of Btk was critical for mast cells to exhibit fully normal degranulation and production/secretion in response to FcεRI stimulation. In accord with this finding, transfection of xid-sBMMCs with a constitutively active form of Btk, Btk* with E41K mutation (60), resulted in the secretion of even higher levels of TNF-α than did transfection of the cells with wt Btk (Fig. 5 A). Interestingly, an SH3 mutant (P265L) could induce at least partial restoration of cytokine producing/secretory capacity. Therefore, an intact SH3 domain does not appear to be required for the expression of at least some Btk function in this system.

Remarkably, we also found that xid (R28C mutation in the PH domain) Btk, when overexpressed, could, depending on the cytokine, partially or apparently fully restore the cytokine producing/secretory capacity in both xid and btk null mast cells. The requirement of Btk domains for degranulation is similar to that for cytokine producing/secretory capacity (Fig. 5, A–C). Interestingly, xid Btk overexpressors released histamine as efficiently as wt Btk transfectants upon FcεRI cross-linking with an optimal concentration of antigen (Fig. 5,C). At first glance, these results appear inconsistent with the finding that xid mast cells express defects in FcεRI-dependent function. However, xid-BMMCs express only 1/5 to 1/3 as much Btk protein as do wt-BMMCs. By contrast, btk null–sBMMCs that had been transfected with xid btk cDNA expressed 20–30% more Btk protein than did cells transfected with wt btk cDNA, and both types of transfectant greatly overexpressed Btk protein relative to levels in xid-BMMCs. Finally, the defects in FcεRI-dependent mast cell function in xid-BMMCs are less severe than those in the btk null–BMMCs; in fact, at optimal concentrations of antigen challenge, xid-BMMCs released histamine at levels that were indistinguishable from those of wt-BMMCs (Fig. 2 A). Taken together with the results obtained with the kinase-dead and SH3 mutants, these results suggest that the kinase activity of Btk is strictly required for Btk function in degranulation and cytokine production and secretion, but that other domains, such as the PH and SH3 domains, are not as essential for these functions of Btk in mast cell activation.

Several Btk-associated molecules have been described. PH domain–binding molecules include phosphatidylinositol 4,5-bisphosphate (and related phosphoinositides; references 6466), the β subunits of GTP-binding proteins (67, 68), PKC (43), and BAP-135 (69). A proline-rich sequence in the Tec homology domain binds SH3 domains of Src family PTKs (70, 71), whereas the SH3 domain of Btk interacts with a protooncogene product, p120c-cbl (72). Differential binding requirements of some of PH domain–associated signaling molecules may account for the observed differences in the biochemical capacities or biological functions of xid versus wt Btk.

We found that wt, but not kinase-dead (K390R), Emt could partially compensate for the absence of Btk in the expression of FcεRI-dependent mast cell degranulation and cytokine production/secretion. Emt protein levels in wt and btk null mast cells, as revealed by immunoblotting with an anti-Emt polyclonal antibody that cross-reacts with Btk, were estimated to be ∼10% of the Btk level in wt mast cells (Fig. 5 D), assuming that the antibody binds to Emt and Btk with the same efficiency. Given this finding and the results of our emt transfection experiments, it is possible that the retention of limited degranulation and cytokine production capacity in xid or btk null mutant mast cells may reflect low-level expression of Emt (and/or other Tec family PTKs) in these cells. This possibility remains to be investigated using mast cells devoid of both Btk and Emt (and/or other Tec family) proteins.

Btk Regulates the Transcription of Several Cytokine Genes.

The abnormalities in the production/secretion of cytokines in btk mutant mast cells seem to be at the transcriptional level. Thus, levels of TNF-α mRNA induced by FcεRI cross-linking in wt mast cells exceeded that in xid mast cells by at least fivefold. This difference in mRNA levels could be due to differences in the transcription rate and/or in the mRNA stability, both phenomena known for IL-2 and other cytokines (73). In addition, the TNF-α mRNA has AU-rich sequences at the 3′ untranslated region that predispose for mRNA degradation and repress its translation (7476). Although the possibilities of differential cytokine mRNA stabilities and differential derepression of mRNA translation between wt and btk mutant mast cells are not ruled out by this study, the notion of Btk-mediated regulation of cytokine gene transcriptions can account largely for our data. Thus, promoter reporter assays using the constructs without the 3′ AU-rich sequences demonstrated significant differences between wt and btk mutant mast cells in the transcriptional activity of the TNF-α and IL-2 promoters and individual cis-elements (see below) upon FcεRI cross-linking. By contrast, we found that the xid mutation had little or no detectable effect on the posttranslational regulation of TNF-α expression. All of these data are consistent with the fact that the expression of this cytokine is regulated at the transcriptional level by the activation of critical transcription factors in activated T and B cells (56– 58, 77).

Similar observations were made with promoter reporter constructs to examine individual cis-acting elements. Thus, NFAT activity was activated by FcεRI stimulation, as previously shown in a mast cell line, CPII (78). However, FcεRI-induced transcriptional activation of an NFAT–luciferase construct was lower in btk null cells than in wt cells. In contrast, NFκB–luciferase and c-fos–luciferase activities were induced (at relatively low levels) by FcεRI stimulation, but the extent of transcriptional activation of these constructs was similar in wt versus btk null mast cells (data not shown). These observations are consistent with the results obtained with btk null mast cells that had been transfected with vector, wt btk, or kinase-dead btk cDNAs (Fig. 4 B). NFATp binds to four sites in the TNF-α promoter (58). Together with NFAT, the CRE site just upstream of κ3, an NFATp-binding site, binds c-Jun and ATF-2 transcription factors to activate this gene in response to TCR/CD3 stimulation (57, 58). NFATp binds to five sites in the IL-2 gene promoter (55) and cooperatively binds with c-Fos– c-Jun heterodimers or c-Jun–c-Jun homodimers at these sites (55, 79). Thus, it is likely that the regulation of the TNF-α and IL-2 genes in mast cells also involves NFAT and AP-1 proteins.

Btk-dependent Signaling Pathways.

Btk is activated by the phosphorylation of tyrosine-551 by Lyn (80). Activated Btk in turn autophosphorylates Tyr-223 in the SH3 domain (81). Chicken DT-40 B lymphoma cells in which the btk gene was knocked out exhibited reduced tyrosine phosphorylation of PLC-γ2 and little [Ca2+]i rise in response to anti-IgM stimulation, suggesting a role of Btk in the regulation of intracellular Ca2+ concentrations through direct or indirect phosphorylation of PLC-γ2 (82). Reaction products of PLC, inositol 1,4,5-trisphosphate, and diacylglycerol, mobilize Ca2+ from intracellular storage sites and activate PKC, respectively (for review see reference 83). Increased Ca2+ concentrations also lead to the activation and nuclear translocation of NFAT by dephosphorylation of cytoplasmic NFAT by the calcium/calmodulin–dependent phosphatase, calcineurin (for review see reference 84).

Our recent data indicates that Btk regulates JNKs and, to a lesser extent, p38, representing two of the three major MAP kinases (i.e., ERKs, JNKs, and p38) that are activated upon FcεRI cross-linking (59). Thus, upon FcεRI cross-linking, xid and btk null mast cells exhibited much reduced JNK activation compared with wt mast cells. Notably, the activities of ERKs upon FcεRI cross-linking were not significantly different between wt and btk mutant mast cells. The activity of phospholipase A2, a key enzyme of arachidonic acid cascade, was shown to be regulated by ERK, which in turn is regulated by Syk in rat basophilic leukemia RBL-2H3 cells (85). Accordingly, the lack of effect of btk mutations on ERK activity after FcεRI cross-linking probably explains our finding that leukotriene levels released from btk null mast cells were similar to those from wt mast cells (data not shown). Similarly, we found that the transcriptional activity of a c-fos–luciferase construct was not affected by btk mutations in mast cells (c-fos is also downstream of ERK).

btk mutations would be expected to impair signaling through JNKs (59). Targets of JNK include the transcription factors c-Jun and ATF-2. JNK phosphorylates the critical residues of the activation domains of these proteins to activate them (8688). c-Jun and ATF-2, in cooperation with NFAT, were shown to bind to the CRE and κ3 sites, respectively, which are required for the induction of the TNF-α promoter (57, 58). In the case of the IL-2 gene, Fos–Jun heterodimers cooperatively bind with NFAT proteins at four of the five NFAT-binding sites in the IL-2 promoter (55). Taken together with these previous findings, our current data are consistent with the hypothesis that Btk can regulate two arms of the FcεRI signaling process, i.e., the PLC/Ca2+/PKC and JNK signaling pathways (Fig. 6).

On the other hand, the regulation of FcεRI signaling is potentially very complex, and not all of these complexities are illustrated in Fig. 6. For example, we found that the secretion of TNF-α and other cytokines upon FcεRI cross-linking was greater in mast cells that had been cultured in the presence of both IL-3 and SCF as compared with that in cells that had been cultured in IL-3 alone. This might reflect, at least in part, the phosphorylation and enzymatic activation of PLC-γ by c-Kit (89, 90), the receptor for SCF. JNK is also activated transiently by SCF stimulation of BMMCs (59). Therefore, SCF/c-Kit–dependent activation of both PLC/Ca2+/PKC and JNK pathways probably contributed to cytokine gene induction in mast cells that were maintained in IL-3 and SCF.

We thank Drs. Ken-ichi Arai, Takashi Yokota, and Lisako Tsuruta for plasmids. We thank Drs. Kimishige Ishizaka, Howard Grey, and Amnon Altman for critical reading of an early version of the manuscript.

1
Galli
SJ
New concepts about the mast cell
N Engl J Med
1993
328
257
265
[PubMed]
2
Beaven
MA
,
Metzger
H
Signal transduction by Fc receptors: the FcεRI case
Immunol Today
1993
14
222
226
[PubMed]
3
Ravetch
JV
,
Kinet
J-P
Fc receptors
Annu Rev Immunol
1991
9
457
492
[PubMed]
4
Reth
M
Antigen receptor tail clue
Nature
1989
338
383
384
[PubMed]
5
Cambier
JC
New nomenclature for the Reth motif (or ARH1/TAM/ARAM/YXXL)
Immunol Today
1995
16
110
[PubMed]
6
Benhamou
M
,
Gutkind
JS
,
Robbins
KC
,
Siraganian
RP
Tyrosine phosphorylation coupled to IgE receptor–mediated signal transduction and histamine release
Proc Natl Acad Sci USA
1990
87
5327
5330
[PubMed]
7
Kawakami
T
,
Inagaki
N
,
Takei
M
,
Fukamachi
H
,
Coggeshall
KM
,
Ishizaka
K
,
Ishizaka
T
Tyrosine phosphorylation is required for mast cell activation by FcεRI cross-linking
J Immunol
1992
148
3513
3519
[PubMed]
8
Stephen
V
,
Benhamou
M
,
Gutkind
JS
,
Robbins
KC
,
Siraganian
RP
FcεRI-induced protein tyrosine phosphorylation of pp72 in rat basophilic leukemia cells (RBL-2H3). Evidence for a novel signal transduction pathway unrelated to G protein activation and phosphatidylinositol hydrolysis
J Biol Chem
1992
267
5434
5441
[PubMed]
9
Hibbs
ML
,
Tarlinton
DM
,
Armes
J
,
Grail
D
,
Hodgson
G
,
Maglitto
R
,
Stacker
SA
,
Dunn
AR
Multiple defects in the immune system of Lyn-defective mice, culminating in autoimmune disease
Cell
1995
83
301
311
[PubMed]
10
Rivera
V
,
Brugge
JS
Clustering of Syk is sufficient to induce tyrosine phosphorylation and release of allergic mediators from rat basophilic leukemia cells
Mol Cell Biol
1995
15
1582
1590
[PubMed]
11
Zhang
J
,
Berenstein
EH
,
Evans
RL
,
Siraganian
RP
Transfection of Syk protein tyrosine kinase reconstitutes high affinity IgE receptor-mediated degranulation in a Syk-negative variant of rat basophilic leukemia RBL-2H3 cells
J Exp Med
1996
184
71
79
[PubMed]
12
Jouvin
M-H
,
Adamczewski
M
,
Numerof
R
,
Letourneur
O
,
Valle
A
,
Kinet
J-P
Differential control of the tyrosine kinases Lyn and Syk by the two signaling chains of the high affinity immunoglobulin E receptor
J Biol Chem
1994
269
5918
5925
[PubMed]
13
Kihara
H
,
Siraganian
RP
Src homlogy 2 domains of Syk and Lyn bind to tyrosine-phosphorylated subunits of the high affinity IgE receptor
J Biol Chem
1994
269
22427
22432
[PubMed]
14
Rowley
RB
,
Burkhardt
AL
,
Chao
H-G
,
Matsueda
GR
,
Bolen
JB
Syk protein-tyrosine kinase is regulated by tyrosine-phosphorylated Igα/Igβ immunoreceptor tyrosine activation motif binding and autophosphorylation
J Biol Chem
1995
270
11590
11594
[PubMed]
15
Shiue
L
,
Zoller
MJ
,
Brugge
JS
Syk is activated by phosphotyrosine-containing peptides representing the tyrosine-based activation motifs of the high affinity receptor for IgE
J Biol Chem
1995
270
10498
10502
[PubMed]
16
Yamada
N
,
Kawakami
Y
,
Kimura
H
,
Fukamachi
H
,
Baier
G
,
Altman
A
,
Kato
T
,
Inagaki
Y
,
Kawakami
T
Structure and expression of novel protein tyrosine kinases, Emb and Emt, in hematopoietic cells
Biochem Biophys Res Commun
1993
192
231
240
[PubMed]
17
Tang
B
,
Mano
H
,
Yi
HT
,
Ihle
JN
Tec kinase associates with c-kit and is tyrosine phosphorylated and activated following stem cell factor binding
Mol Cell Biol
1994
14
8432
8437
[PubMed]
18
Kawakami
Y
,
Yao
L
,
Tsukada
S
,
Witte
ON
,
Kawakami
T
Tyrosine phosphorylation and activation of Bruton tyrosine kinase upon FcεRI cross-linking
Mol Cell Biol
1994
14
5108
5113
[PubMed]
19
Kawakami
Y
,
Yao
L
,
Tashiro
M
,
Gibson
S
,
Mills
GB
,
Kawakami
T
Activation and interaction with protein kinase C of a cytoplasmic tyrosine kinase, Itk/Tsk/ Emt, upon FcεRI cross-linking on mast cells
J Immunol
1995
155
3556
3562
[PubMed]
20
Eiseman
E
,
Bolen
JB
Engagement of the high-affinity IgE receptor activates srcprotein-related tyrosine kinases
Nature
1992
355
78
80
[PubMed]
21
Hutchcroft
JE
,
Geahlen
RL
,
Deanin
GG
,
Oliver
JM
FcεRI-mediated tyrosine phosphorylation and activation of the 72-kDa protein-tyrosine kinase, PTK72, in RBL-2H3 rat tumor mast cells
Proc Natl Acad Sci USA
1992
89
9107
9111
[PubMed]
22
Benhamou
M
,
Ryba
NJP
,
Kihara
H
,
Nishikata
H
,
Siraganian
RP
Protein-tyrosine kinase p72sykin high affinity IgE receptor signaling. Identification as a component of pp72 and association with the receptor γ chain after receptor aggregation
J Biol Chem
1993
268
23318
23324
[PubMed]
23
Tsukada
S
,
Saffran
D
,
Rawlings
DJ
,
Parolini
O
,
Allen
RC
,
Klisak
I
,
Sparkes
RS
,
Kubagawa
H
,
Mohandas
T
,
Quan
S
et al
Deficient expression of a B cell cytoplasmic tyrosine kinase in human X-linked agammaglobulinemia
Cell
1993
72
279
290
[PubMed]
24
Vetrie
D
,
Vorechovsky
I
,
Sideras
P
,
Holland
J
,
Davies
A
,
Flinter
F
,
Hammarstrom
L
,
Kinnon
C
,
Levinsky
R
,
Bobrow
M
et al
The gene involved in X-linked agammaglobulinemia is a member of the src family of protein-tyrosine kinases
Nature
1993
361
226
233
[PubMed]
25
Thomas
JD
,
Sideras
P
,
Smith
CIE
,
Vorechovsky
I
,
Chapman
V
,
Paul
WE
Colocalization of X-linked agammaglobulinemia and X-linked immunodeficiency genes
Science
1993
261
355
358
[PubMed]
26
Rawlings
DJ
,
Saffran
DC
,
Tsukada
S
,
Largaespada
DA
,
Grimaldi
JC
,
Cohen
L
,
Mohr
RN
,
Bazan
JF
,
Howard
M
,
Copeland
NG
et al
Mutation of unique region of Bruton's tyrosine kinase in immunodieficient XID mice
Science
1993
261
358
361
[PubMed]
27
Aoki
Y
,
Isselbacher
KJ
,
Pillai
S
Bruton tyrosine kinase is tyrosine phosphorylated and activated in pre-B lymphocytes and receptor-ligated B cells
Proc Natl Acad Sci USA
1994
91
10606
10609
[PubMed]
28
Saouaf
SJ
,
Mahajan
S
,
Rowley
RB
,
Kut
SA
,
Fargnoll
J
,
Burkhardt
AL
,
Tsukada
S
,
Witte
ON
,
Bolen
JB
Temporal differences in the activation of three classes of non-transmembrane protein tyrosine kinases following B-cell antigen receptor surface engagement
Proc Natl Acad Sci USA
1994
91
9524
9528
[PubMed]
29
de Weers
M
,
Brouns
GS
,
Hinshelwood
S
,
Kinnon
C
,
Schuurman
RKB
,
Hendriks
RW
,
Borst
J
B-cell antigen receptor stimulation activates the human Bruton's tyrosine kinase, which is deficient in X-linked agammaglobulinemia
J Biol Chem
1994
269
23857
23860
[PubMed]
30
Santos-Argumedo
L
,
Lund
FE
,
Heath
AW
,
Solvason
N
,
Mu
WW
,
Grimaldi
JC
,
Parkhouse
RME
,
Howard
M
CD38 unresponsiveness of xid B cells implicates Bruton's tyrosine kinase (btk)as a regulator of CD38 induced signal transduction
Int Immunol
1995
7
163
170
[PubMed]
31
Yamashita
Y
,
Miyake
K
,
Kikuchi
Y
,
Takatsu
K
,
Noda
S
,
Kosugi
A
A monoclonal antibody against a murine CD38 homologue delivers a signal to B cells for prolongation of survival and production against apoptosis in vitro: unresponsiveness of X-linked immunodeficient B cells
Immunology
1995
85
248
255
[PubMed]
32
Hasbold
J
,
Klaus
GGB
B cells from CBA/N mice do not proliferate following ligation of CD40
Eur J Immunol
1994
24
152
157
[PubMed]
33
Sato
S
,
Katagiri
T
,
Takaki
S
,
Kikuchi
Y
,
Hitoshi
Y
,
Yonehara
S
,
Tsukada
S
,
Kitamura
D
,
Watanabe
T
,
Witte
ON
,
Takatsu
K
IL-5 receptor–mediated tyrosine phosphorylation of SH2/SH3-containing proteins and activation of Bruton's tyrosine and Janus 2 kinases
J Exp Med
1994
180
2101
2111
[PubMed]
34
Matsuda
T
,
Takahashi-Tezuka
M
,
Fukuda
T
,
Okuyama
Y
,
Fujitani
Y
,
Tsukada
S
,
Mano
H
,
Hirai
H
,
Witte
ON
,
Hirano
T
Association and activation of Btk and Tec tyrosine kinases by gp130, a signal transducer of the interleukin-6 family of cytokines
Blood
1995
85
627
633
[PubMed]
35
Go
NF
,
Castle
BE
,
Barrett
R
,
Kastelein
R
,
Dang
W
,
Mosmann
TR
,
Moor
KW
,
Howard
M
Interleukin 10, a novel B cell stimulatory factor: unresponsiveness of X-chromosome–linked immunodeficiency B cells
J Exp Med
1990
172
1625
1631
[PubMed]
36
August
A
,
Gibson
S
,
Kawakami
Y
,
Kawakami
T
,
Mills
GB
,
Dupont
B
CD28 is associated with and induces the immediate tyrosine phosphorylation and activation of the Tec family kinase ITK/EMT in the human Jurkat leukemic T-cell line
Proc Natl Acad Sci USA
1994
91
9347
9351
[PubMed]
37
Gibson
S
,
August
A
,
Kawakami
Y
,
Kawakami
T
,
Dupont
B
,
Mills
GB
The EMT/ITK/TSK (EMT) tyrosine kinase is activated during TCR signaling
J Immunol
1996
156
2716
2722
[PubMed]
38
Liao
XC
,
Littman
DR
Altered T cell receptor signaling and disrupted T cell development in mice lacking Itk
Immunity
1995
3
757
769
[PubMed]
39
Khan
WN
,
Alt
FW
,
Gerstein
RM
,
Malynn
BA
,
Larsson
I
,
Rathbun
G
,
Davidson
L
,
Mueller
S
,
Kantor
AB
,
Herzenberg
LA
et al
Defective B cell development and function in Btk-deficient mice
Immunity
1995
3
283
299
[PubMed]
40
Kerner
JD
,
Appleby
MW
,
Mohr
RN
,
Chien
S
,
Rawlings
DJ
,
Maliszewski
CR
,
Witte
ON
,
Perlmutter
RM
Impaired expansion of mouse B cell progenitors lacking Btk
Immunity
1995
3
301
312
[PubMed]
41
Inagaki
N
,
Goto
S
,
Nagai
H
,
Koda
A
Homologous passive cutaneous anaphylaxis in various strains of mice
Int Arch Allergy Appl Immunol
1986
81
58
62
[PubMed]
42
Nagai
H
,
Sakurai
T
,
Inagaki
N
,
Mori
H
An immunopharmacological study of the biphasic allergic skin reaction
Biol Pharm Bull
1995
18
239
245
[PubMed]
43
Yao
L
,
Kawakami
Y
,
Kawakami
T
The pleckstrin homology domain of Btk tyrosine kinase interacts with protein kinase C
Proc Natl Acad Sci USA
1994
91
9175
9179
[PubMed]
44
Gibson
S
,
Leung
B
,
Squire
JA
,
Hill
M
,
Arima
N
,
Gross
P
,
Hogg
D
,
Mills
GB
Identification, cloning, and characterization of a novel human T-cell–specific tyrosine kinase located at the hematopoietin complex on chromosome 5q
Blood
1993
83
1561
1572
[PubMed]
45
Horton
RM
,
Hunt
HD
,
Ho
SN
,
Pullen
JK
,
Pease
LR
Engineering hybrid genes without the use of restriction enzymes: gene splicing by overlap extension
Gene
1989
77
61
68
[PubMed]
46
Onishi
M
,
Kinoshita
S
,
Morikawa
Y
,
Shibuya
A
,
Phillips
J
,
Lanier
LL
,
Gorman
DM
,
Nolan
GP
,
Miyajima
A
,
Kitamura
T
Applications of retrovirus-mediated expression cloning
Exp Hematol
1996
24
324
329
[PubMed]
47
Pear
WS
,
Nolan
GP
,
Scott
ML
,
Baltimore
D
Production of high-titer helper-free retroviruses by transient transfection
Proc Natl Acad Sci USA
1993
90
8392
8396
[PubMed]
48
Siraganian
RP
An automated continuous-flow system for the extraction and fluorometric analysis of histamine
Anal Biochem
1974
57
383
394
[PubMed]
49
Tsuruta
L
,
Lee
H-J
,
Masuda
ES
,
Koyano-Nakagawa
N
,
Arai
N
,
Arai
K
,
Yokota
T
Cyclic AMP inhibits expresssion of the IL-2 gene through the nuclear factor of activated T cells (NF-AT) site, and transfection of NF-AT cDNAs abrogates the sensitivity of EL-4 cells to cyclic AMP
J Immunol
1995
154
5255
5264
[PubMed]
50
Deng
T
,
Karin
M
c-Fos transcriptional activity stimulated by H-Ras–activated protein kinase distinct from JNK and ERK
Nature
1994
371
171
175
[PubMed]
51
Wershil
BK
,
Wang
Z-S
,
Gordon
JR
,
Galli
SJ
Recruitment of neutrophils during IgE-dependent cutaneous late phase reactions in the mouse is mast cell–dependent. Partial inhibition of the reaction with antiserum against tumor necrosis factor–α
J Clin Invest
1991
87
446
453
[PubMed]
52
Inagaki
N
,
Goto
S
,
Yamasaki
M
,
Nagai
H
,
Koda
A
Studies on vascular permeability increasing factors involved in 48-hour homologous PCA in the mouse ear
Int Arch Allergy Appl Immunol
1986
80
285
290
[PubMed]
53
Kriegler
M
,
Perez
C
,
DeFay
K
,
Albert
I
,
Lu
SD
A novel form of TNF/cachectin is a cell surface cytotoxic transmembrane protein: ramifications for the complex physiology of TNF
Cell
1988
53
45
53
[PubMed]
54
Shaw
J-P
,
Utz
PJ
,
Durand
DB
,
Toole
JJ
,
Emmel
EA
,
Crabtree
GR
Identification of a putative regulator of early T cell activation genes
Science
1988
241
202
205
[PubMed]
55
Rooney
JW
,
Sun
Y-L
,
Glimcher
LH
,
Hoey
T
Novel NFAT sites that mediate activation of the interleukin-2 promoter in response to T-cell receptor stimulation
Mol Cell Biol
1995
15
6299
6310
[PubMed]
56
Goldfeld
AE
,
McCaffrey
PG
,
Strominger
JL
,
Rao
A
Identification of a novel cyclosporin-sensitive element in the human tumor necrosis factor α gene promoter
J Exp Med
1993
178
1365
1379
[PubMed]
57
Tsai
EY
,
Jain
J
,
Pesavento
PA
,
Rao
A
,
Goldfeld
AE
Tumor necrosis factor alpha gene regulation in activated T cells involves ATF-2/Jun and NFATp
Mol Cell Biol
1996
16
459
467
[PubMed]
58
Tsai
EY
,
Yie
J
,
Thanos
D
,
Goldfeld
AE
Cell-type–specific regulation of the human tumor necrosis factor alpha gene in B cells and T cells by NFATp and ATF-2/ JUN
Mol Cell Biol
1996
16
5232
5244
[PubMed]
59
Kawakami
Y
,
Miura
T
,
Bissonnette
R
,
Hata
D
,
Khan
WN
,
Kitamura
T
,
Maeda-Yamamoto
M
,
Hartman
SE
,
Yao
L
,
Alt
FW
,
Kawakami
T
Bruton's tyrosine kinase regulates apoptosis and JNK/SAPK kinase activity
Proc Natl Acad Sci USA
1997
94
3938
3942
[PubMed]
60
Li
T
,
Tsukada
S
,
Satterthwaite
A
,
Havlik
MH
,
Park
H
,
Takatsu
K
,
Witte
ON
Activation of Bruton's tyrosine kinase (BTK) by a point mutation in its pleckstrin homology (PH) domain
Immunity
1995
2
1
20
[PubMed]
61
Clark
SG
,
Stern
MJ
,
Horvitz
HR
C. elegans cell-signalling gene sem-5 encodes a protein with SH2 and SH3 domains
Nature
1992
356
340
344
[PubMed]
62
Conley
ME
,
Parolini
O
,
Rohrer
J
,
Campana
D
X-linked agammaglobulinemia: new approaches to old questions based on the identification of the defective gene
Immunol Rev
1994
138
5
21
[PubMed]
63
Scher
I
The CBA/N mouse strain: an experimental model illustrating the influence of X chromosome on immunity
Adv Immunol
1982
33
1
71
[PubMed]
64
Harlan
JE
,
Hajduk
PJ
,
Yoon
HS
,
Fesik
SW
Pleckstrin homology domains bind to phosphatidylinositol-4,5-bisphosphate
Nature
1994
371
168
170
[PubMed]
65
Salim
K
,
Bottomley
MJ
,
Querfurth
E
,
Zvelebil
MJ
,
Gout
I
,
Scaife
R
,
Margolis
RL
,
Gigg
R
,
Smith
CIE
,
Driscoll
PC
et al
Distinct specificity in the recognition of phosphoinositides by the pleckstrin homology domains of dynamin and Bruton's tyrosine kinase
EMBO (Eur Mol Biol Organ) J
1996
15
6241
6250
[PubMed]
66
Fukuda
M
,
Kojima
T
,
Kabayama
H
,
Mikoshiba
K
Mutation of the pleckstrin homology domain of Bruton's tyrosine kinase in immunodeficiency impaired inositol 1,3,4,5-terakisphosphate binding capacity
J Biol Chem
1996
271
30303
30306
[PubMed]
67
Touhara
K
,
Inglese
J
,
Pitcher
JA
,
Shaw
G
,
Lefkowitz
RJ
Binding of G protein βγ-subunits to pleckstrin homology domains
J Biol Chem
1994
269
10217
10220
[PubMed]
68
Wang
D-S
,
Shaw
R
,
Winkelmann
JC
,
Shaw
G
Binding of PH domains of β-adrenergic receptor kinase and β-spectrin to WD40/β-transducin repeat containing regions of the β-subunit of trimeric G-proteins
Biochem Biophys Res Commun
1994
203
29
35
[PubMed]
69
Yang
W
,
Desiderio
S
BAP-135, a target for Bruton's tyrosine kinase in response to B cell receptor engagement
Proc Natl Acad Sci USA
1997
94
604
609
[PubMed]
70
Cheng
G
,
Ye
Z-S
,
Baltimore
D
Binding of Bruton's tyrosine kinase to Fyn, Lyn, or Hck through a Src homology 3 domain–mediated interaction
Proc Natl Acad Sci USA
1994
91
8152
8155
[PubMed]
71
Yang
W
,
Malek
SN
,
Desiderio
S
An SH3-binding site conserved in Bruton's tyrosine kinase and related tyrosine kinases mediates specific protein interactions in vitro and in vivo
J Biol Chem
1995
270
20832
20840
[PubMed]
72
Cory
GOC
,
Lovering
RC
,
Hinshelwood
S
,
MacCarthy-Morrogh
L
,
Levinsky
RJ
,
Kinnon
C
The protein product of the c-cblprotooncogene is phosphorylated after B cell receptor stimulation and binds the SH3 domain of Bruton's tyrosine kinase
J Exp Med
1995
182
611
615
[PubMed]
73
Lindsten
T
,
June
CH
,
Ledbetter
JA
,
Stella
G
,
Thompson
CB
Regulation of lymphokine messenger RNA stability by a surface-mediated T cell activation pathway
Science
1989
244
339
343
[PubMed]
74
Caput
D
,
Beutler
B
,
Hartog
K
,
Thayer
R
,
Brown-Shimer
S
,
Cerami
A
Identification of a common nucleotide sequence in the 3′-untranslated region of mRNA molecules specifying inflammatory mediators
Proc Natl Acad Sci USA
1986
83
1670
1674
[PubMed]
75
Beutler
B
,
Krochin
N
,
Milsark
IW
,
Luedke
C
,
Cerami
A
Control of cachectin (tumor necrosis factor) synthesis: mechanisms of endotoxin resistance
Science
1986
232
977
980
[PubMed]
76
Han
J
,
Brown
T
,
Beutler
B
Endotoxin-responsive sequences control cachectin/tumor necrosis factor biosynthesis at the translational level
J Exp Med
1990
171
465
475
[PubMed]
77
Goldfeld
AE
,
Tsai
EY
,
Kincaid
R
,
Belshaw
PJ
,
Schreiber
SL
,
Strominger
JL
,
Rao
A
Calcineurin mediates human tumor necrosis factor α gene induction in stimulated T and B cells
J Exp Med
1994
180
763
768
[PubMed]
78
Prieschl
EE
,
Pendl
GG
,
Harrer
NE
,
Baumruker
T
p21raslinks FcεRI to NF-AT family member in mast cells. The AP3-like factor in this cell type is an NF-AT family member
J Immunol
1995
155
4963
4970
[PubMed]
79
Jain
J
,
McCaffrey
PG
,
Miner
Z
,
Kerppola
TK
,
Lambert
JN
,
Verdine
GL
,
Curran
T
,
Rao
A
The T-cell transcription factor NFATp is a substrate for calcineurin and interacts with Fos and Jun
Nature
1993
365
352
355
[PubMed]
80
Rawlings
DJ
,
Scharenberg
AM
,
Park
H
,
Wahl
MI
,
Lin
S
,
Kato
RM
,
Fluckiger
A-C
,
Witte
ON
,
Kinet
J-P
Activation of BTK by a phosphorylation mechanism initiated by SRC family kinases
Science
1996
271
822
825
[PubMed]
81
Park
H
,
Wahl
MI
,
Afar
DEH
,
Turck
CW
,
Rawlings
DJ
,
Tam
C
,
Scharenberg
AM
,
Kinet
J-P
,
Witte
ON
Regulation of Btk function by a major autophosphorylation site within the SH3 domain
Immunity
1996
4
515
525
[PubMed]
82
Takata
M
,
Kurosaki
T
A role for Bruton's tyrosine kinase in B cell antigen receptor–mediated activation of phospholipase C-γ2
J Exp Med
1996
184
31
40
[PubMed]
83
Nishizuka
Y
The role of protein kinase C in cell surface signal transduction and tumour promotion
Nature
1984
308
693
698
[PubMed]
84
Rao
A
NF-ATp: a transcription factor required for the co-ordinate induction of several cytokine genes
Immunol Today
1994
15
274
281
[PubMed]
85
Hirasawa
N
,
Scharenberg
AM
,
Yamamura
H
,
Beaven
MA
,
Kinet
J-P
A requirement for Syk in the activation of the microtubule-associated protein kinase/phopholipase A2pathway by FcεRI is not shared by a G protein–coupled receptor
J Biol Chem
1995
270
10960
10967
[PubMed]
86
Gupta
S
,
Campbell
D
,
Derijard
B
,
Davis
RJ
Transcription factor ATF2 regulation by the JNK signal transduction pathway
Science
1995
267
389
393
[PubMed]
87
Livingstone
C
,
Patel
G
,
Jones
N
ATF-2 contains a phosphorylation-dependent transcriptional activation domain
EMBO (Eur Mol Biol Organ) J
1995
14
1785
1797
[PubMed]
88
van Dam
H
,
Wilhelm
D
,
Herr
I
,
Steffen
A
,
Herrlich
P
,
Angel
P
ATF-2 is preferentially activated by stress-activated protein kinases to mediate c-juninduction in response to genotoxic agents
EMBO (Eur Mol Biol Organ) J
1995
14
1798
1811
[PubMed]
89
Lev
S
,
Givol
D
,
Yarden
Y
A specific combination of substrates is involved in signal transduction by the kit-encoded receptor
EMBO (Eur Mol Biol Organ) J
1991
10
647
654
[PubMed]
90
Reith
AD
,
Ellis
C
,
Lyman
SD
,
Anderson
DM
,
Williams
DE
,
Bernstein
A
,
Pawson
T
Signal transduction by normal isoforms and Wmutant variants of the Kit receptor tyrosine kinase
EMBO (Eur Mol Biol Organ) J
1991
10
2451
2459
[PubMed]

1Abbreviations used in this paper: BMMC, bone marrow–derived cultured mast cells; Btk, Bruton's tyrosine kinase; ITAM, immunoreceptor tyrosine–based activation motif; NFAT, nuclear factor of activated T cells; PCA, passive cutaneous anaphylactic; PH, pleckstrin homology; PKC, protein kinase C; PLC, phospholipase C; PTK, protein tyrosine kinase; SCF, stem cell factor; SH, Src homology.

This study was partly supported by National Institutes of Health grants RO1 AI-33617 and RO1 AI-38348 (T. Kawakami) and R37 AI-23990 and RO1 CA-72074 (S.J. Galli) and is Publication No. 147 from the La Jolla Institute for Allergy and Immunology.

Author notes

Address correspondence to Toshiaki Kawakami, La Jolla Institute for Allergy and Immunology, 10355 Science Center Dr., San Diego, CA 92121. Phone: 619-558-3500; Fax: 619-558-3526; E-mail: toshi_ kawakami@liai.org