The transcription factor T-bet was identified in CD4+ T cells, and it controls interferon γ production and T helper type 1 cell differentiation. T-bet is expressed in certain other leukocytes, and we recently showed (Lord, G.M., R.M. Rao, H. Choe, B.M. Sullivan, A.H. Lichtman, F.W. Luscinskas, and L.H. Glimcher. 2005. Blood. 106:3432–3439) that it regulates T cell trafficking. We examined whether T-bet influences homing of mast cell progenitors (MCp) to peripheral tissues. Surprisingly, we found that MCp homing to the lung or small intestine in T-bet−/− mice is reduced. This is reproduced in adhesion studies using bone marrow–derived MCs (BMMCs) from T-bet−/− mice, which showed diminished adhesion to mucosal addresin cellular adhesion molecule–1 (MAdCAM-1) and vascular cell adhesion molecule–1 (VCAM-1), endothelial ligands required for MCp intestinal homing. MCp, their precursors, and BMMCs do not express T-bet, suggesting that T-bet plays an indirect role in homing. However, adoptive transfer experiments revealed that T-bet expression by BM cells is required for MCp homing to the intestine. Furthermore, transfer of WT BM-derived dendritic cells (DCs) to T-bet−/− mice restores normal MCp intestinal homing in vivo and MCp adhesion to MAdCAM-1 and VCAM-1 in vitro. Nonetheless, T-bet−/− mice respond vigorously to intestinal infection with Trichinella spiralis, eliminating a role for T-bet in MC recruitment to sites of infection and their activation and function. Therefore, remarkably, T-bet expression by DCs indirectly controls MCp homing to mucosal tissues.

Mast cell progenitors (MCp) are derived from the hematopoietic stem cells present in the BM and develop along the myelomonocytic pathway. The exact pathway is still somewhat controversial, described in C57BL/6 mice as either via a basophil/MC bipotent progenitor found in the spleen or derived via an alternate path in BM (1, 2). Regardless, MCp transit through the circulation, immigrate into peripheral tissues, and mature into tissue MCs under the influence of the local microenvironment (35). Within the peripheral tissues, MCs exhibit different phenotypes according to their tissue location, with distinctive histochemical staining characteristics and differences in the protease and proteoglycan content of their secretory granules (68). They are broadly classified as connective tissue–type MCs, which are found in connective tissues throughout the body, often adjacent to blood microvessels and in the peritoneal cavity of mice and rats, where they have been clearly implicated in the innate immune response to bacteria (9, 10), and mucosal MCs, which appear in mucosal regions within the lung and the intestine. Both connective tissue–type MCs and mucosal MCs act as effector cells during inflammation and play an important role in allergic inflammation of the airways and in the intestinal immune response to helminth infection (11, 12).

Previous studies have proposed a model of basal homing of MCp to the intestine (13, 14). In this tissue, MCp homing is a constitutive, dynamic process that appears to be independent of adaptive immunity. The model envisions the regulation of MCp trafficking at the level of integrin CD49d/β7 interactions, with its counterreceptors mucosal addresin cellular adhesion molecule–1 (MAdCAM-1) and vascular cell adhesion molecule–1 (VCAM-1) expressed by vascular endothelium (13). Furthermore, the CXC chemokine receptor (CXCR) 2 provides promigratory signals critical to MCp trafficking to the intestine (14, 15).

T-bet is a T-box transcription factor essential to the generation and function of Th1 cells, and its deficiency leads to predominantly Th2-mediated responses (16). T-bet expression has been identified in other leukocytes, including NK cells, DCs, and CD8+ cells, where it directs strong transactivation of IFN-γ transcripts (1719). We previously identified a critical role for T-bet in CD4+ T cell trafficking via its ability to regulate selectin-dependent adhesion and surface expression of CXCR3 (20), and more recently T-bet has also been identified as a regulator of NKT cell trafficking (21). We do not know of any studies of the role of T-bet in trafficking of other leukocyte types. In this paper, we investigate the role of T-bet in MCp trafficking and report the unexpected finding that MCp homing to the mucosal tissues of the lung and the intestine is impaired in T-bet−/− mice. This striking observation led us to investigate the hypothesis that T-bet expression by MCp controls recruitment and homing to mucosal tissues. We found that there is no expression of T-bet in MCs at several stages of development. Yet, adoptive transfer of T-bet–deficient BM into sublethally irradiated WT mice still results in impaired homing, demonstrating for the first time that another radiation sensitive BM-derived cell plays an essential role in MCp homing to mucosal tissues. Furthermore, we have shown that this BM-derived cell is most likely a DC, because adoptive transfer of WT BM-derived DCs (BMDCs) into T-bet−/− mice reconstitutes MCp intestinal homing, and co-culture of WT BMDCs with T-bet−/− BM-derived MCs (BMMCs) restored the binding of BMMCs to MAdCAM-1 and VCAM-1 in vitro to normal levels.

MCp homing to the mucosal tissues is impaired in T-bet−/− mice

We have recently shown that CD4+ T cell trafficking in T-bet−/− mice is dramatically reduced (20). Our interest in MC trafficking then led us to investigate whether MCp homing to the mucosal tissues is impaired in T-bet−/− mice. We measured the total number of MCp in the intestine and the lung, as well as in the BM and the spleen. T-bet deficiency resulted in a nearly total loss of the MCp reservoir normally present in the small intestine and in a significant decrease in the lung, whereas MCp numbers in the spleen and the BM were comparable to those in WT mice. This finding was consistently observed in both BALB/c (Fig. 1 A) and C57BL/6 (Fig. 1 B) mice strains lacking T-bet. Consistent with previous findings (13), the number of MCp found in the tissues examined in the BALB/c strain was greater than that in C57/BL6 mice. Histochemical analysis of tissue sections prepared from intestine and spleen of adult, age-matched T-bet−/− and WT mice also showed that fewer MCs were detected in the intestine of T bet−/− compared with WT mice. In the spleen, which showed no differences in MCp, MCs were also diminished in the T-bet−/− mice (Fig. 2). These data indicate that under basal conditions in the absence of T-bet there is a tissue-specific homing defect in MCp homing to mucosal tissues.

T-bet−/− MCs exhibit impaired binding to VCAM-1 and MAdCAM-1 under flow conditions

Having observed a lack of homing of MCp to the intestine, which is the largest reservoir of committed MCp in the mouse (22), we postulated that T-bet−/− MCp might exhibit diminished α4 integrin–dependent adhesive interactions with endothelial cell adhesion molecules VCAM-1 and MAdCAM-1, because these endothelial ligands have previously been shown to be essential for MCp homing to the intestine (13). To assess this further, we measured adhesion of WT and T-bet−/− BMMCs to immobilized, recombinant mouse VCAM-1 or MAdCAM-1 under conditions of physiological laminar shear flow. Because the number of MCp in the BM or in circulating blood of adult mice is low (1, 23), it is impossible to isolate quantities sufficient to perform these adhesion assays. Therefore, we and others (24, 25) have used BMMCs generated in vitro as a surrogate model to study the different properties and adhesion pathways of MCs. T-bet−/− BMMCs displayed markedly reduced binding to both VCAM-1 and MAdCAM-1 across a range of shear flow (Fig. 3). Incubation of WT and T -bet−/− BMMCs with blocking antibodies against the α4 integrin chain abolished BMMC binding to VCAM-1 (unpublished data). These data suggest that T-bet affects the adhesive interactions between MCp and intestinal endothelial VCAM-1 and MAdCAM-1. Because MCp express receptors for keratinocyte-derived chemokine (KC/CXC chemokine ligand [CXCL] 1) and chemokines coimmobilized with adhesion molecules are known to trigger leukocyte stable arrest under shear flow conditions (26, 27), we examined whether this or other chemokines (stromal cell–derived factor 1α [SDF-1α]) coimmobilized with VCAM-1 or MAdCAM-1 would overcome the adhesion defect. Neither WT nor T-bet−/− MCp showed an increase in adhesion (Fig. S1, A–C) nor resistance to detachment after adhesion to MAdCAM-1 (Fig. S1 D), suggesting that MCs and T cells do not exhibit the same recognition response in this assay. We next tested whether surface expression of α4 integrins (CD49d), which bind to VCAM-1 and MAdCAM-1, were altered. FACS analysis revealed very low levels of surface expression of α4 and β7 integrins and the chemokine receptor CXCR2 in BMMCs generated from either WT or T-bet−/− mice (Fig. S2), in agreement with previous reports (24, 28, 29). Thus, the defect is not explained by a loss of α4β7 integrin expression or a lack of chemokine-induced arrest under flow conditions.

T-bet is not expressed in BMMCs or MCp

Because this defect in adhesion suggests a direct role for T-bet in controlling MCp adhesion, the next set of studies determined T-bet expression in cultured BMMCs or other cells along the developmental pathway that we have previously described (1). Western blot analysis revealed that T-bet is not expressed in either T-bet−/− or WT BMMCs under basal conditions or after stimulation with nonphysiologic (PMA + ionomycin) or physiologic (LPS and IgE cross-linking) stimuli (Fig. 4 A). In contrast, T-bet was easily detected in T cytotoxic type 1 cells (Tc1), analyzed in parallel as a positive control for T-bet expression.

We next considered the possibility that T-bet was not expressed in BMMCs because of a loss of expression under the culture conditions required for successful in vitro differentiation. Because previous studies have reported that the Lin/c-kit+/Sca1 CD34+ cell population from adult BM gives rise to MCs (1, 30), we used a high-speed FACS approach on the BM of WT adult mice to isolate these cells and probe the isolated cells for T-bet expression by quantitative real-time PCR, using Th1 cells as a positive control (Fig. 4 B). T-bet was present in the WT BM, but at very low levels. The cell populations sorted from the BM as Lin or Lin+ cells, which correspond to uncommitted or committed BM progenitors (of MCs and other lineages), respectively, also expressed very low levels of T-bet (Fig. 4 B, inset plotted with expanded y axis). The expression of T-bet by whole BM cells is most likely caused by Lin+ cells, because T-bet was below detectable levels in the population (Lin) that leads to MCp production. We also tested for the expression of T-bet in other hematopoietic progenitors present in the BM and committed to the myeloid pathway, such as the common myeloid progenitor, the granulocyte monocyte progenitor (GMP), the recently described basophil MC progenitor (BMCP) (1), and the basophil progenitor (BaP), as well as in MCp derived in vitro from GMP and mature peritoneal MCs. None of these cells, progenitors or mature MCs, expressed T-bet. In contrast, T bet was easily detected in positive-control Th1 cells (Fig. 4 C). We conclude that T-bet is not expressed in committed MCp, its immediate precursors, or BMMCs derived in vitro, and hence postulate that T-bet plays an indirect role in the homing of MCp to the mucosal tissues.

Reconstitution of MCp in sublethally irradiated BM reconstituted (SIBR) mice with BM cells from T-bet−/− or BALB/c mice

In view of the inability of our assays to detect T-bet expression in MCp or cultured BMMCs, we were forced by the observed striking defect of MCp pools in certain mucosal tissues of T-bet–null mice to pursue other approaches to understanding how T-bet exerts this effect. To define whether the observed homing defect was due exclusively to a defect in the T-bet−/− BM (including MCp or other BM precursors expressing T-bet) or, alternatively, to a defect of the local intestinal microenvironment by itself, we depleted the hematopoietic precursors in WT mice by sublethal irradiation and then reconstituted the animals by adoptive transfer of either WT or T-bet−/− BM cells (SIBR). The number of MCp found in the intestine of WT-SIBR mice reconstituted with T-bet−/− BM was very low compared with that in WT-SIBR mice reconstituted with WT BM. In contrast, the numbers of MCp in the spleen and BM were not significantly different in WT mice reconstituted with T bet−/− BM from those reconstituted with WT BM. (Fig. 5 A). On the other hand, sublethally irradiated T-bet−/− mice reconstituted with BM from WT mice showed total reconstitution of intestinal MCp, as well as of BM and spleen MCp (Fig. 5 B). These results demonstrate that the MCp that arise from the T bet−/− BM show a defect in homing to the intestine despite normal numbers of precursors in the spleen or BM, whereas the MCp arising from the WT BM and transferred into a T-bet−/− host show adequate tissue homing of MCp, including homing to the intestine. These data implicate a defect or loss of T-bet–expressing cells in the BM compartment that is responsible for the lack of MCp adhesion and intestinal-specific homing in the T-bet−/− mice and rule out the possibility of a defect in the intestinal compartment of T-bet−/− mice.

T-bet expressed in BMDCs regulates MCp intestinal homing via VCAM-1 and MAdCAM-1 ligands

Because the findings in Fig. 5 demonstrated that a defect in the BM compartment of T-bet−/− mice is responsible for the lack of MCp homing to the intestine, we performed experiments to identify the BM cell type responsible. Based on the literature (18), DCs are good candidates because their development, differentiation, and activation is independent of T-bet, but DCs can up-regulate T-bet and secrete IFN-γ (18). We derived DCs from WT BM by culture in GM-CSF for 7 d and transferred these BMDCs into WT or T-bet−/− mice. Strikingly, T–bet−/− mice given WT BMDCs showed levels of MCp in the intestine comparable to WT mice given BMDCs in parallel when evaluated 14 d later. Transfer of BMDCs into WT mice did not significantly increase the number of MCp (Fig. 6 A). In contrast, T-bet−/− animals that did not receive BMDCs exhibited a dramatic reduction of MCp in the intestine as compared with WT mice.

The role of T-bet+ BMDCs in controlling MCp intestinal homing was further investigated by testing whether co-culture of WT BMDCs with T-bet−/− BMMCs restored T-bet BMMC binding to VCAM-1 and MAdCAM-1. Indeed, a 6-h co-culture of T-bet−/− BMMCs with WT BMDCs completely rescued binding to VCAM-1 and MAdCAM-1, as these cells exhibited no binding under conditions of shear flow that was comparable to WT BMMCs (Fig. 6, B and C). BMMC adhesion was abolished by anti–α4 integrin antibody. These data strongly suggest that T-bet in the DCs plays a role in homing of MCp to the intestine by regulating the α4β7-dependent binding to the endothelial cell ligands VCAM-1 and MAdCAM-1, which are necessary for the appropriate homing of MCp to the intestine (13).

T-bet−/− mice are able to mount an immune response to Trichinella spiralis infection

Infection with T. spiralis stimulates a large increase in the number of MCs residing in the small intestine (8). The rapid expulsion of T. spiralis is associated with and dependent on the pronounced mastocytosis, which is turn is mediated by a Th2-type response (3134). Lack of the intestinal MCs and MCp in β7 integrin–deficient mice resulted in a delayed MC hyperplasia and delayed rejection of the T. spiralis from infected mice (35). Thus, we assessed the intestinal MC response of WT and T-bet−/− mice to infection with T. spiralis. Despite the low number of resident MCs and reduced MCp reservoir in the intestine of T-bet−/− mice, a robust mucosal MC response was noted in the T-bet–deficient mice. The response was even greater than that observed in WT mice infected in parallel and resulted in an enhanced elimination of the worms from the intestine (Fig. 7) regardless of the strain background. The enhanced clearance of infection was observed with T-bet−/− mice in a Th2 cell–type background strain (Fig. 7 A, BALB/c mice) and with T-bet−/− mice in a Th1 cell–type background strain (Fig. 7 B, C57BL/6 mice). The local MC hyperplasia in response to the infection was also markedly increased in the T-bet−/− mice, even more so than in WT mice (Fig. 7, C and D). These results are consistent with the view that the T-bet–null mouse is polarized toward the induction of a Th2 cell inflammatory milieu (19), and T. spiralis, which provides a potent stimulus, provokes a more vigorous MC proliferation response to this pathogen.

We report on the unexpected and novel finding that the transcription factor T-bet expressed in DCs indirectly regulates MCp homing to mucosal tissues. The involvement of T-bet in Th1 T cell polarization, IFN-γ production by DCs, and terminal maturation of NK cells has been documented (1619), whereas the role for T-bet in leukocyte trafficking is an emerging area. Recent reports showed that T-bet was necessary for trafficking of CD4+ T cells (20), CD8+ T cells (36), and NKT cells (21) to sites of immune reactions. Our interest in understanding the factors that control MC trafficking (1, 3) led us to study the effect of T-bet deficiency on MCp homing to tissues.

The initial observation of a dramatically lower number of MCp in mucosal tissues in T-bet−/− animals (Fig. 1), specifically in the small intestine as measured by limiting dilution bioassay, was very surprising. That this reduction in MCp was physiologically relevant was corroborated by the observed 50–70% reduction in mature tissue MCs, as detected by histological analysis of the small intestine and spleen (Fig. 2). As some mature MCs in the spleen have been shown to be derived from the intestine, at least after infection with T. spiralis (37), the decrease in splenic MC numbers could reflect the decreased numbers of intestinal MCs in the T-bet−/− mice or a maturational defect within the spleen.

From earlier studies, we knew that T-bet was necessary for both the biosynthesis of functional selectin ligands and the expression of chemokine receptors in CD4+ T cells (20), which guided us to consider that MCp adhesion by itself was defective. Because the hematopoietic pool of committed MCp in mice is estimated to be <30,000 cells (Fig. 1) (1, 23), we used BMMCs cultured in vitro as a surrogate for MCp cells to study their adhesive properties under physiological shear flow conditions. These experiments revealed that T-bet–deficient BMMCs adhered poorly to both VCAM-1 and MAdCAM-1 (Fig. 3). Furthermore, coimmobilized chemokines such as SDF-1α/CXCL12 or KC/CXCL1 were not sufficient to reconstitute or augment adhesion. Despite the decreased adhesion, no loss of α4β7 integrin or CXCR2 expression was detected (Fig. S2), perhaps implicating a defect in α4 integrin–dependent or chemokine receptor signaling.

At this juncture, the picture emerging was that T-bet expression in MCp selectively controlled homing to mucosal tissues (but not development in BM or spleen) via the α4β7 integrin–VCAM-1/MAdCAM-1/CXCR2 adhesion pathway. An unexpected finding was that T-bet transcripts were below detectable levels in mature MCs isolated from the peritoneal cavity, in BMMCs or in committed intestinal MCp obtained by immunoisolation and high-speed FACS sorting (Fig. 4). We did detect T-bet expression in Lin+ BM cells, which are precursors of many other cell types, but not in the uncommitted Lin population or in other early myeloid progenitors, such us common myeloid progenitor, GMP, and BMCP, or the committed progeny of the BMCP, the MCp or BaP. From these collective findings, we concluded that T-bet cannot directly regulate MCp homing via α4β7 integrin/CXCR2–dependent adhesion. To further understand the role of T-bet, we addressed whether the defect resided in other cellular components of the BM and/or in the intestinal milieu by reconstituting sublethally irradiated WT or T-bet−/−–null animals with BM from WT or T-bet−/− mice. These studies indicated that the defect lies in the BM compartment and not in the intestinal microenvironment (Fig. 5), because T-bet–null mice reconstituted with normal BM cells had similar numbers of intestinal MCp to WT mice reconstituted with WT BM cells. In contrast, WT mice reconstituted with T-bet−/− BM cells showed decreased MCp homing to the intestine. Because the genotype of the BM was critical and not that of the intestine, these results indicate that T-bet expression in BM cells, other than the MCp or its immediate precursors, is critical to proper homing of MCp to tissues. Although surprising, this is similar to the recent observation that TNF-α production by CD11b+ BM cells was critical to development and expansion of BMMCs (38) and supports the concept that MC development along the myeloid pathway requires interaction with other leukocytes. Given the restricted expression of T-bet within leukocytes of the BM, the most likely T-bet+ cell found in this environment is the DC.

We have tested this hypothesis by adoptive transfer of WT DCs into T-bet−/− mice and found that these animals now exhibited normal homing of MCp to intestine. Furthermore, T-bet−/− BMMCs co-cultured with WT DCs recover the ability to bind to MAdCAM-1 and VCAM-1 at levels that are comparable to WT BMMCs. We conclude that the presence of DCs expressing T-bet in the BM compartment is required for normal α4β7 integrin–dependent binding to MAdCAM-1 and VCAM-1 and MCp intestinal homing.

We further probed whether such a defect in the MCp pool increased the susceptibility of these animals to a serious outcome when challenged with a helminth infection. Based on studies in β7 integrin–deficient mice, which lack MCp and MCs in the intestine and show delayed rejection of an intestinal helminth, we anticipated a similar finding in T-bet–null mice, even though these mice are polarized toward Th2 responses (35, 13). Unexpectedly, the rejection of the worms by T-bet mice was equal to or even better than that by WT mice (Fig. 7), consistent with previous reports of an association of a rapid expulsion of T. spiralis with a pronounced mastocytosis mediated by a Th2-type response and the findings that T-bet–null mice are Th2 cell polarized (19, 35). Our finding suggests that like in the β7 integrin–null mice, the adhesion defect in α4β7 integrin/CXCR2–mediated signaling for homing is compensated by other pathways elicited by the potent Th2 cell–mediated immune response that overcomes the initial dependence on the former molecules.

We have yet to identify the specific DC T-bet–dependent mechanisms in BM that are responsible for the defect in the α4β7 integrin–CXCR2 adhesion pathway. We do not suspect that reduced α4 integrin/CXCR2 expression is responsible, because BMMCs derived from WT and T-bet–deficient mice showed equivalent, albeit low, levels of surface-expressed α4 and β7 integrins and CXCR2. The loss of the α4 integrin during protracted in vitro culture that we observed (unpublished data) has also been reported previously (28, 29). Another possibility is that T-bet−/− cells have a defect in α4 integrin or CXCR2 signaling that leads to the observed loss of adhesion. We note that one should interpret these results cautiously, because BMMCs are not MCp; they are only a tool, the best in vitro surrogate cells currently available for studying MCp.

T-bet controls a multitude of genes in several cell types, making it difficult to discern how T-bet is indirectly controlling MCp homing to mucosal tissues. Our experiments provide evidence that the transcription factor T-bet expressed in DCs within the BM indirectly imprints a program in developing MCp that ensures appropriate homing to the mucosal tissues. Regardless of the fact that the IFN-γ gene is one of the main direct targets of T-bet (16, 18), we were not able to increase the MCp intestinal homing in T-bet−/− mice by intraperitoneal injection of IFN-γ, and more strikingly, the MCp present in the WT intestine were depleted after IFN-γ treatment, probably due to apoptosis of MCs induced by IFN-γ, as described by other authors (unpublished data) (39). Thus, it seems most likely that IFN-γ is not directly responsible for MCp homing and that some other factor causes the homing defect in T-bet–deficient mice. Nonetheless, these findings are remarkable, as it is the first indication that another innate effector cell directly influences MC development in a way that affects its trafficking in the absence of overt inflammation.

In summary, these observations provide further evidence that the transcription factor T-bet is involved in leukocyte trafficking and show, for the first time, a role for T-bet expression in DCs for the constitutive homing of MCp cells to mucosal tissues.

Reagents.

Dulbecco's PBS and RPMI 1640 were purchased from Invitrogen. The following reagents were obtained from the indicated sources: mouse MAdCAM-1 chimeric protein (T. Yednock, Elan Corporation, San Francisco, CA), recombinant mouse VCAM-1-Fc chimera (R&D Systems), and recombinant mouse IL-3, stem cell factor (SCF), SDF-1α, GM-CSF, and KC/CXCL1 (PeproTech). The monoclonal antibodies to mouse PSGL-1 (clone 2PH1), CD62L (MEL-14), α4 integrin (clones R1-2 and 9C10), CXCR4 (clone 2B11), MHC class II (anti–H-2Kd; SF1-1.1–FITC), and CD11c (HL3-APC) were purchased from BD Biosciences; anti–mouse CXCR2 was an anti-peptide antibody made in rabbits, as previously described (14). Anti–T-bet antibody (clone 4B10) was obtained from Santa Cruz Biotechnology, Inc. IgE-DNP, PMA, ionomycin, LPS, fetal calf serum, and polyclonal antibody to β-actin were purchased from Sigma-Aldrich. Gr1 (anti-Ly6G/C; RB6-8C5–PE) was obtained from eBioscience.

Mice.

Previously described 6–8-wk-old WT and T-bet−/− mice (19) on C57/BL6 and BALB/c backgrounds were purchased from the Jackson Laboratory. Mice were maintained in a specific pathogen-free barrier unit at the Harvard New Research Building facilities or at the Dana Farber Cancer Institute animal facilities. All experimental protocols were reviewed and approved by the Institutional Animal Care and Use Committee in accordance with policies of the Public Health Service.

MC isolation and cell culture.

BM MCs were generated as previously detailed (7). 0.5 × 106 BMMCs/ml were cultured for 4 wk in T-25 culture flasks (Corning) under standard conditions in the presence of 10 ng/ml IL-3 and 10 ng/ml SCF.

BMDC isolation and cell culture.

BMDCs were generated as previously detailed (40, 41). BMDCs from WT mice were cultured in the presence of 10ng/ml GM-CSF, with supplementation on days 0, 3, and 6. On day 7, they were analyzed by FACS. The BMDC purity was 65–75% CD11c+, class II+, and Gr1, with contaminating cells predominantly Gr-1+ cells (Fig. S3). This Gr-1 population is likely to contain a mixture of mature granulocytes, monocytes, and macrophages, none of which express T-bet, as previously described (18). 2 × 106 cultured BMDCs/mouse were injected intravenously into T-bet−/− mice. Also on day 7, they were stimulated with 10 U/ml IFN-γ for 1 h and co-cultured with BMMCs (DC/MC = 1:1.5) for 6 h. These co-cultures were tested for adhesion to VCAM-1 and MAdCAM-1 under shear flow conditions.

Adhesion assays under physiological shear flow conditions.

Glass coverslips (25-mm diameter; Carolina Biological Supply) were coated with 10 μg/ml VCAM-1 or 20 μg/ml MAdCAM-1. BMMC adhesive interactions were examined under conditions of fluid shear stress using a video microscopy system, as previously described (42). Cell accumulation was determined after the initial minute of each flow rate by counting the number of adherent cells in four different fields using a 20× phase-contrast objective (Plan Apo; Nikon) and recorded with videomicroscopy connected to VideoLab software (Ed Marcus Laboratories). Data are the mean ± SEM (n = 3 experiments). P ≤ 0.5 or less was considered statistically significant using the paired t test or one-way analysis of variance for multiple groups.

Preparation of tissue mononuclear cells (MNCs) and MCp limiting dilution assay.

The procedure for determining the number of MCp was essentially as previously described (13, 22). Individual tissues from two mice were pooled and processed to isolate MNCs. MNCs were serially diluted in 96-well flat-bottom microtiter plates, and 100 μl of γ-irradiated (30 Gy) spleen-derived feeder cells in media containing 20 ng/ml IL-3 and 20 ng/ml SCF were added. MNCs were cultured for 12 d before MC colonies were identified and counted with inverted microscopes (CK, Olympus; TMS, Nikon. The MCp concentration is expressed as MCp/tissue and is derived by multiplying the concentration of MCp per 106 MNCs by the MNC yield/organ. For the BM, the number represents the total MCp per femur.

T-bet expression by Western blot.

BMMCs from WT mice were stimulated for 24 h with 2 μg/ml LPS, 50 ng/ml PMA plus 2 μM ionomycin, or IgE cross-linking (1μg/ml of mouse αDNP-IgE for 16 h plus 10 ng/ml DNP-OVA). Immunoblot analysis was performed by using the 4B10 mAb, as previously described (16).

Sorting of MCp, real-time PCR, and semiquantitative RT-PCR.

Sorting of myeloid progenitors was as previously described (30). Myeloid progenitors were sorted as Lin Sca-1 c-kit+ CD34+, as previously described (43). BaP or MCp sorting was as previously described (1). Total RNA was extracted from the sorted cells or from in vitro–generated mouse Th1 cells or Tc1 cells (provided by N. Grabie, Brigham and Women's Hospital, Boston, MA) as positive controls using TRIzol solution (Invitrogen) and reverse transcribed using a cDNA synthesis kit (iScript; Bio-Rad Laboratories). The amount of amplicon generated was monitored with an ABI Prism 7700 (Applied Biosystems). A specific probe labeled with both reporter and a quencher dye was added into the Taqman PCR mix at the beginning of the reaction. The cycle number was normalized to β-actin.

For semiquantitative PCR, the following primers were used to amplify T-bet and β-actin: T-bet (forward), 5′-GCCAGGGAACCGCTTATATGTC-3′; T-bet (reverse), 5′-CTGTGAGATCATATCCTTGGGCTG-3′; β-actin (forward), 5′-GTGGGCCGCTCTAGGCACCA-3′; and β-actin (reverse), 5′-CGGTTGGCCTTAGGGTTCAGGGGGG-3′.

Immunohistochemistry.

Thin sections (1.5-μm thickness) of tissue were fixed with 4% paraformaldehyde and subjected to the chloroacetate esterase reactivity cytochemistry procedure for MC histology analysis, as previously described (8). The results are expressed as the number of MCs per cross-sectional area of the intestine or spleen.

T. spiralis infection and worm burden determination.

Trichinella larvae were obtained as detailed previously (8). Mice were infected with ∼450 larvae suspended in 200 μl PBS by direct gastric installation. Worm burden was determined after 13 d, as previously described (44). Each experimental group comprised six animals.

Sublethal irradiation with BM reconstitution.

Animals were irradiated (GammaCell-40; Atomic Energy of Canada) for 4.5 min, 5 gray (500 rad). 2 h after irradiation, mice were given intravenous injections of 107 BM cells that had been isolated from femurs and tibias of syngenic or congenic animals. 14 d after irradiation, mice were killed, and MCp limiting dilution assays were performed.

Statistical analysis.

Data are the mean ± SEM (n = 5 separate experiments with two mice per group in each experiment), unless indicated otherwise. P ≤ 0.05 was considered statistically significant by the paired t test or one-way analysis of variance for multiple groups using Prism software (version 4; GraphPad).

Online supplemental material.

The online supplemental material contains additional information about adhesion studies under flow conditions, as well as characterization of DCs and BMMCs. Fig. S1 evaluates the adhesion of BMMCs under shear flow conditions to VCAM-1 and MAdCAM-1, coimmobilized with the chemokines KC and SDF-1α. Fig. S2 shows expression of CXCR2 and α4 and β7 integrins in BMMCs by FACS analysis. Fig. S3 shows the phenotypes of the DCs used in our assays.

We wish to thank Dr. Ravi Rao for his helpful discussions regarding T-bet function in MC biology, Dr. Nir Grabie for his gift of mouse Tc1 lymphocytes, Dr. Gail Newton for help with designing primers for T-bet RT-PCR, and Drs. Daniel Friend and Yuhui Xu for MC histological analysis.

These studies were supported by grants from the Fulbright-Spanish Ministry of Education and Science (to P. Alcaide), the P.E. Lindahl Fund from the Royal Swedish Academy of Sciences (to J. Hallgren), the National Institutes of Health (HL36028, HL53993, and HL56985 to F.W. Luscinskas; CA112663 and AI56296 to L.H. Glimcher; and HL 036110 and AI 031599 to M.F. Gurish), and the Ellison Medical Foundation (L.H. Glimcher).

The authors have no conflicting financial interests.

1
Arinobu, Y., H. Iwasaki, M.F. Gurish, S. Mizuno, H. Shigematsu, H. Ozawa, D.G. Tenen, K.F. Austen, and K. Akashi.
2005
. Developmental checkpoints of the basophil/mast cell lineages in adult murine hematopoiesis.
Proc. Natl. Acad. Sci. USA.
102
:
18105
–18110.
2
Chen, C.C., M.A. Grimbaldeston, M. Tsai, I.L. Weissman, and S.J. Galli.
2005
. Identification of mast cell progenitors in adult mice.
Proc. Natl. Acad. Sci. USA.
102
:
11408
–11413.
3
Rodewald, H.R., M. Dessing, A.M. Dvorak, and S.J. Galli.
1996
. Identification of a committed precursor for the mast cell lineage.
Science.
271
:
818
–822.
4
Williams, C.M., and S.J. Galli.
2000
. The diverse potential effector and immunoregulatory roles of mast cells in allergic disease.
J. Allergy Clin. Immunol.
105
:
847
–859.
5
Metcalfe, D.D., D. Baram, and Y.A. Mekori.
1997
. Mast cells.
Physiol. Rev.
77
:
1033
–1079.
6
Enerback, L.
1966
. Mast cells in rat gastrointestinal mucosa. 2. Dye-binding and metachromatic properties.
Acta Pathol. Microbiol. Scand.
66
:
303
–312.
7
Gurish, M.F., N. Ghildyal, H.P. McNeil, K.F. Austen, S. Gillis, and R.L. Stevens.
1992
. Differential expression of secretory granule proteases in mouse mast cells exposed to interleukin 3 and c-kit ligand.
J. Exp. Med.
175
:
1003
–1012.
8
Friend, D.S., N. Ghildyal, K.F. Austen, M.F. Gurish, R. Matsumoto, and R.L. Stevens.
1996
. Mast cells that reside at different locations in the jejunum of mice infected with Trichinella spiralis exhibit sequential changes in their granule ultrastructure and chymase phenotype.
J. Cell Biol.
135
:
279
–290.
9
Echtenacher, B., D.N. Mannel, and L. Hultner.
1996
. Critical protective role of mast cells in a model of acute septic peritonitis.
Nature.
381
:
75
–77.
10
Malaviya, R., T. Ikeda, E. Ross, and S.N. Abraham.
1996
. Mast cell modulation of neutrophil influx and bacterial clearance at sites of infection through TNF-alpha.
Nature.
381
:
77
–80.
11
Trotter, C.M., and T.S. Orr.
1973
. A fine structure study of some cellular components in allergic reactions. 1. Degranulation of human mast cells in allergic asthma and perennial rhinitis.
Clin. Allergy.
3
:
411
–425.
12
Ruitenberg, E.J., and A. Elgersma.
1976
. Absence of intestinal mast cell response in congenitally athymic mice during Trichinella spiralis infection.
Nature.
264
:
258
–260.
13
Gurish, M.F., H. Tao, J.P. Abonia, A. Arya, D.S. Friend, C.M. Parker, and K.F. Austen.
2001
. Intestinal mast cell progenitors require CD49dβ7 (α4β7 integrin) for tissue-specific homing.
J. Exp. Med.
194
:
1243
–1252.
14
Abonia, J.P., K.F. Austen, B.J. Rollins, S.K. Joshi, R.A. Flavell, W.A. Kuziel, P.A. Koni, and M.F. Gurish.
2005
. Constitutive homing of mast cell progenitors to the intestine depends on autologous expression of the chemokine receptor CXCR2.
Blood.
105
:
4308
–4313.
15
Nilsson, G., J.A. Mikovits, D.D. Metcalfe, and D.D. Taub.
1999
. Mast cell migratory response to interleukin-8 is mediated through interaction with chemokine receptor CXCR2/Interleukin-8RB.
Blood.
93
:
2791
–2797.
16
Szabo, S.J., S.T. Kim, G.L. Costa, X. Zhang, C.G. Fathman, and L.H. Glimcher.
2000
. A novel transcription factor, T-bet, directs Th1 lineage commitment.
Cell.
100
:
655
–669.
17
Townsend, M.J., A.S. Weinmann, J.L. Matsuda, R. Salomon, P.J. Farnham, C.A. Biron, L. Gapin, and L.H. Glimcher.
2004
. T-bet regulates the terminal maturation and homeostasis of NK and Valpha14i NKT cells.
Immunity.
20
:
477
–494.
18
Lugo-Villarino, G., R. Maldonado-Lopez, R. Possemato, C. Penaranda, and L.H. Glimcher.
2003
. T-bet is required for optimal production of IFN-gamma and antigen-specific T cell activation by dendritic cells.
Proc. Natl. Acad. Sci. USA.
100
:
7749
–7754.
19
Szabo, S.J., B.M. Sullivan, C. Stemmann, A.R. Satoskar, B.P. Sleckman, and L.H. Glimcher.
2002
. Distinct effects of T-bet in TH1 lineage commitment and IFN-gamma production in CD4 and CD8 T cells.
Science.
295
:
338
–342.
20
Lord, G.M., R.M. Rao, H. Choe, B.M. Sullivan, A.H. Lichtman, F.W. Luscinskas, and L.H. Glimcher.
2005
. T-bet is required for optimal proinflammatory CD4+ T-cell trafficking.
Blood.
106
:
3432
–3439.
21
Matsuda, J.L., Q. Zhang, R. Ndonye, S.K. Richardson, A.R. Howell, and L. Gapin.
2006
. T-bet concomitantly controls migration, survival, and effector functions during the development of Valpha14i NKT cells.
Blood.
107
:
2797
–2805.
22
Guy-Grand, D., M. Dy, G. Luffau, and P. Vassalli.
1984
. Gut mucosal mast cells. Origin, traffic, and differentiation.
J. Exp. Med.
160
:
12
–28.
23
Sonoda, T., T. Ohno, and Y. Kitamura.
1982
. Concentration of mast-cell progenitors in bone marrow, spleen, and blood of mice determined by limiting dilution analysis.
J. Cell. Physiol.
112
:
136
–140.
24
Boyce, J.A., E.A. Mellor, B. Perkins, Y.C. Lim, and F.W. Luscinskas.
2002
. Human mast cell progenitors use alpha4-integrin, VCAM-1, and PSGL-1 E-selectin for adhesive interactions with human vascular endothelium under flow conditions.
Blood.
99
:
2890
–2896.
25
Kitamura, Y., T. Kasugai, N. Arizono, and H. Matsuda.
1993
. Development of mast cells and basophils: processes and regulation mechanisms.
Am. J. Med. Sci.
306
:
185
–191.
26
Cinamon, G., and R. Alon.
2003
. A real time in vitro assay for studying leukocyte transendothelial migration under physiological flow conditions.
J. Immunol. Methods.
273
:
53
–62.
27
Rao, R.M., T.V. Betz, D.J. Lamont, M.B. Kim, S.K. Shaw, R.M. Froio, F. Baleux, F. Arenzana-Seisdedos, R. Alon, and F.W. Luscinskas.
2004
. Elastase release by transmigrating neutrophils deactivates endothelial-bound SDF-1α and attenuates subsequent T lymphocyte transendothelial migration.
J. Exp. Med.
200
:
713
–724.
28
Fehlner-Gardiner, C.C., S. Uniyal, C.G. von Ballestrem, and B.M. Chan.
1996
. Differential utilization of VLA-4 (alpha 4 beta 1) and -5 (alpha 5 beta 1) integrins during the development of mouse bone marrow-derived mast cells.
Differentiation.
60
:
317
–325.
29
Ducharme, L.A., and J.H. Weis.
1992
. Modulation of integrin expression during mast cell differentiation.
Eur. J. Immunol.
22
:
2603
–2607.
30
Iwasaki-Arai, J., H. Iwasaki, T. Miyamoto, S. Watanabe, and K. Akashi.
2003
. Enforced granulocyte/macrophage colony-stimulating factor signals do not support lymphopoiesis, but instruct lymphoid to myelomonocytic lineage conversion.
J. Exp. Med.
197
:
1311
–1322.
31
Finkelman, F.D., T. Shea-Donohue, J. Goldhill, C.A. Sullivan, S.C. Morris, K.B. Madden, W.C. Gause, and J.F. Urban Jr.
1997
. Cytokine regulation of host defense against parasitic gastrointestinal nematodes: lessons from studies with rodent models.
Annu. Rev. Immunol.
15
:
505
–533.
32
Grencis, R.K., L. Hultner, and K.J. Else.
1991
. Host protective immunity to Trichinella spiralis in mice: activation of Th cell subsets and lymphokine secretion in mice expressing different response phenotypes.
Immunology.
74
:
329
–332.
33
Urban, J., H. Fang, Q. Liu, M.J. Ekkens, S.J. Chen, D. Nguyen, V. Mitro, D.D. Donaldson, C. Byrd, R. Peach, et al.
2000
. IL-13-mediated worm expulsion is B7 independent and IFN-gamma sensitive.
J. Immunol.
164
:
4250
–4256.
34
Urban, J.F., Jr., L. Schopf, S.C. Morris, T. Orekhova, K.B. Madden, C.J. Betts, H.R. Gamble, C. Byrd, D. Donaldson, K. Else, and F.D. Finkelman.
2000
. Stat6 signaling promotes protective immunity against Trichinella spiralis through a mast cell- and T cell-dependent mechanism.
J. Immunol.
164
:
2046
–2052.
35
Artis, D., N.E. Humphreys, C.S. Potten, N. Wagner, W. Muller, J.R. McDermott, R.K. Grencis, and K.J. Else.
2000
. Beta7 integrin-deficient mice: delayed leukocyte recruitment and attenuated protective immunity in the small intestine during enteric helminth infection.
Eur. J. Immunol.
30
:
1656
–1664.
36
Taqueti, V.R., N. Grabie, R. Colvin, H. Pang, P. Jarolim, A.D. Luster, L.H. Glimcher, and A.H. Lichtman.
2006
. T-bet controls pathogenicity of CTLs in the heart by separable effects on migration and effector activity.
J. Immunol.
177
:
5890
–5901.
37
Friend, D.S., M.F. Gurish, K.F. Austen, J. Hunt, and R.L. Stevens.
2000
. Senescent jejunal mast cells and eosinophils in the mouse preferentially translocate to the spleen and draining lymph node, respectively, during the recovery phase of helminth infection.
J. Immunol.
165
:
344
–352.
38
Wright, H.V., D. Bailey, M. Kashyap, C.L. Kepley, M.S. Drutskaya, S.A. Nedospasov, and J.J. Ryan.
2006
. IL-3-mediated TNF production is necessary for mast cell development.
J. Immunol.
176
:
2114
–2121.
39
Mann-Chandler, M.N., M. Kashyap, H.V. Wright, F. Norozian, B.O. Barnstein, S. Gingras, E. Parganas, and J.J. Ryan.
2005
. IFN-gamma induces apoptosis in developing mast cells.
J. Immunol.
175
:
3000
–3005.
40
Inaba, K., M. Inaba, N. Romani, H. Aya, M. Deguchi, S. Ikehara, S. Muramatsu, and R.M. Steinman.
1992
. Generation of large numbers of dendritic cells from mouse bone marrow cultures supplemented with granulocyte/macrophage colony-stimulating factor.
J. Exp. Med.
176
:
1693
–1702.
41
Nolan, K.F., V. Strong, D. Soler, P.J. Fairchild, S.P. Cobbold, R. Croxton, J.A. Gonzalo, A. Rubio, M. Wells, and H. Waldmann.
2004
. IL-10-conditioned dendritic cells, decommissioned for recruitment of adaptive immunity, elicit innate inflammatory gene products in response to danger signals.
J. Immunol.
172
:
2201
–2209.
42
Lim, Y.C., L. Henault, A.J. Wagers, G.S. Kansas, F.W. Luscinskas, and A.H. Lichtman.
1999
. Expression of functional selectin ligands on Th cells is differentially regulated by IL-12 and IL-4.
J. Immunol.
162
:
3193
–3201.
43
Akashi, K., D. Traver, T. Miyamoto, and I.L. Weissman.
2000
. A clonogenic common myeloid progenitor that gives rise to all myeloid lineages.
Nature.
404
:
193
–197.
44
Gurish, M.F., P.J. Bryce, H. Tao, A.B. Kisselgof, E.M. Thornton, H.R. Miller, D.S. Friend, and H.C. Oettgen.
2004
. IgE enhances parasite clearance and regulates mast cell responses in mice infected with Trichinella spiralis.
J. Immunol.
172
:
1139
–1145.

Abbreviations used: BaP, basophil progenitor; BMDC, BM-derived DC; BMCP, basophil MC progenitor; BMMC, BM-derived MC; CXCL and CXCR, CXC chemokine ligand and receptor, respectively; GMP, granulocyte monocyte progenitor; KC, keratinocyte-derived chemokine; MAdCAM-1, mucosal addresin cellular adhesion molecule–1; MC, mast cell; MCp, MC progenitor(s); MNC, mononuclear cell; PMC, peritoneal MC; SCF, stem cell factor; SDF-1α, stromal cell–derived factor 1α; SIBR, sublethally irradiated BM reconstituted; Tc1, T cytotoxic type 1; VCAM-1, vascular cell adhesion molecule–1.

P. Alcaide, T.G. Jones, and G.M. Lord contributed equally to this work.

Supplementary data