TNFR1 is the primary signaling receptor that initiates the majori

TNFR1 is the primary signaling receptor that initiates the majority of inflammatory responses classically attributed to TNF. In contrast, TNFR2 is important in modulating TNFR1-mediated signaling by inducing the depletion of TNF receptor-associated factor 2 (TRAF2) and cellular

inhibitor of apoptosis1 (c-IAP1) proteins and accelerates TNFR1-dependent activation of caspase-8 12, 13. TNFR superfamily members can be classified into two main groups, death domain (DD)-containing receptors such as TNFR1, and TRAF-binding receptors such as TNFR2 that lack a DD 1, 2. Signaling via TNFR1 can have two outcomes. After binding of TNF, TNFR1 recruits the DD-containing adaptor molecule TNFR1-associated DD protein, which functions as a platform to recruit additional signaling molecules for the assembly of alternative Tanespimycin signaling complexes. One complex involves receptor-interacting protein and TRAF2

which links ligand-induced signaling to the activation of the transcription factors NF-κB and AP1 14–17. Another signaling complex is formed dependent on the internalization of activated TNF/TNFR1 complexes. During endocytosis FADD and caspase-8 are recruited to form the death inducing Dorsomorphin signaling complex resulting in TNF-induced apoptosis 2, 14, 15. In this study, we investigated the impact of TNFR2 on regulating cell death or survival as a result of TNFR1 signaling. We tested the hypothesis that in the absence of TNFR2, signaling via TNFR1 would promote cell survival by promoting NF-κB activation by the following mechanism. It is known Resveratrol that TNFR2 signaling leads to the degradation of TRAF2 13. We postulated that in TNFR2-deficient cells, TRAF2 degradation is prevented and the relatively high intracellular levels of TRAF2 in these cells would promote TNFR1-induced NF-κB activation and cell survival. Our results support

this hypothesis. We showed that blocking TNFR2 signaling in anti-CD3+IL-2-activated WT CD8+ T cells resulted in elevated intracellular TRAF2 levels and an increase in their resistance to AICD. Furthermore, blocking anti-TNF-α antibodies significantly reduced TRAF2 accumulation in activated TNFR2−/− CD8+ T cells and increased their susceptibility to AICD. We found that AICD-resistant cells expressed elevated level of phosphorylated IκBα and higher DNA binding activity of the p65 NF-κB subunit, providing further support of our hypothesis that TNFR1 functions as a pro-survival receptor in TNFR2-deficient CD8+ T cells. The activation and differentiation of T cells are dependent on TCR-antigen interaction and the engagement of multiple molecules on the APC by receptors on the T cell. Previously, we demonstrated that TNFR2 not only lowers the threshold for T-cell activation but also provides early costimulatory signals during T-cell activation 6–8.

The difference of plasma sRAGE between patients with normal

The difference of plasma sRAGE between patients with normal Selleckchem Dasatinib (>90 ml/min per 1.73 m2) and lower eGFR was not statistical significant (887.7 ± 82.5 pg/ml versus 949.5±155.1 pg/ml, P = 0.733). The positive rates for ANA, anti-dsDNA, AnuA, anti-Sm were 92.2% (95/103), 53.9% (55/102), 55.7% (54/97), 37.1% (30/89), respectively, in patients with SLE. There was no significant difference between sRAGE levels in patients

with negative ANA and those with different levels of ANA (Fig. 4A). In addition, there was no significant difference between the sRAGE levels in autoantibody-positive patients and those in autoantibody-negative patients (Fig. 4B,C,D). In patients

with SLE, plasma sRAGE levels was negatively correlated with the leucocyte count (n = 95, r = −0.326, P = 0.001, Fig. 5A), absolute values of lymphocytes (n = 95, r = −0.357, P = 0.000, Fig. 5B), neutrophils (n = 95, r = −0.272, P = 0.008, Fig. 5C) and monocytes (n = 95, r = −0.286, P = 0.005, Fig. 5D) in peripheral blood. In this study, we found that plasma sRAGE level in patients with SLE was lower than that in HC, while there was no significant difference of sRAGE level between active and inactive patients. Decreased sRAGE levels in patients with SLE may be explained by the consumption of this soluble receptor. Renard et al. [36] postulated that sRAGE-ligand complexes were eliminated from the blood via spleen and/or liver. Staurosporine solubility dmso It has been demonstrated that the level of HMGB1, one important RAGE ligand, is increased in the acetylcholine circulation of SLE [19, 20], leading to the binding and consumption of sRAGE during the inflammatory process. It is also possible that sRAGE levels in patients with SLE may be regulated by alternative splicing and proteinases and this possibility needs to be clarified in the

future research. sRAGE might not only function as a decoy to exert their inhibitory effects on RAGE, but also act in a more direct way, e.g. binding to cell surface RAGE to block the formation of homodimers [28]. Therefore, decreased levels of sRAGE, which may contribute to enhanced RAGE-mediated pro-inflammatory signalling [27], support the essential role of RAGE in SLE pathology. Our results were different from the recent report showing that blood sRAGE levels in patients with SLE were higher than those in HC and compared with quiescent SLE, blood sRAGE levels are significantly increased during active disease [34]. One explanation for this discrepancy is that use of medication might influence the results. The discrepancy may also be caused by the low number of cases included in that study (only 10 cases of patients with SLE).

These data indicate the critical role of B cells not only for aut

These data indicate the critical role of B cells not only for autoantibody production, but also for CD4+ T cell priming as professional antigen-presenting cells. B cells are therefore an ideal therapeutic target in terms

of not only lowering activities of pathogenic antibodies, but also dampening pathogenic autoimmune responses per se in autoimmune diseases. However, B cell KO mice have a serious problem, in that these mice have major qualitative and quantitative abnormalities in the immune system [7,8]. By contrast, B cell depletion may be a feasible approach to study the function of B cells in autoimmune diseases. Indeed monoclonal antibodies to B cell-specific cell surface molecules such as CD19, CD20, CD79 and to a B cell-surviving factor (B cell lymphocyte stimulator, BLyS) have been used successfully DAPT cost to deplete B cells in vivo and to treat numerous autoimmune and malignant haematopoietic diseases in humans and mice [2,9,10]. Transient depletion of B cells by these means can distinguish between the role of B cells during immune development and during immune responses. CD20 is a B cell-specific

molecule that is expressed on the cell surface during the transition of pre-B to immature B cells but is lost upon plasma cell differentiation [11]. In human autoimmune diseases, rituximab, a chimeric anti-human Erastin concentration CD20 monoclonal antibody, has proved to be effective for treatment of autoimmune diseases, including rheumatoid arthritis, SLE, idiopathic thrombocytopenic purpura, haemolytic anaemia and pemphigus vulgaris [12]. In addition, preliminary clinical studies have shown the therapeutic efficacy of rituximab in a small fraction of Graves’ patients with mild hyperthyroidism [13–16]. In mice, anti-mouse CD20 monoclonal antibodies (anti-mCD20 mAbs) which efficiently eliminate mouse B cells in vivo have been isolated recently

[11,17], and used to treat mouse models of autoimmune thyroiditis, systemic sclerosis, collagen- or proteoglycan-induced http://www.selleck.co.jp/products/BAY-73-4506.html arthritis, Sjögren’s syndrome, SLE and type 1 diabetes [17–22]. Moreover, the soluble decoy receptor-Fc fusion proteins to block B cell surviving factors [BLyS and a proliferation-inducing ligand (APRIL)] reduced TSAb activities and thyroxine (T4) levels in a mouse model of Graves’ disease [23]. In the present study, we evaluated the efficacy of anti-mCD20 mAb in a mouse model of Graves’ disease we have established previously [23]. We found that this approach depleted B cells efficiently and that B cell depletion by this agent was effective for preventing Graves’ hyperthyroidism. Our results indicate the requirement of antibody production and T cell activation by B cells in the early phase of disease initiation for the disease pathogenesis. Female BALB/c mice (6 weeks old) were purchased from Charles River Japan Laboratory Inc. (Tokyo, Japan) and were kept in a specific pathogen-free facility.

[1, 2] Moreover, the allergen-specific CD4+ T cells of non-allerg

[1, 2] Moreover, the allergen-specific CD4+ T cells of non-allergic subjects were mostly either unpolarized or produced low levels of interferon-γ (IFN-γ) and interleukin-10 (IL-10).[1, 2] In the current study, we sought to confirm these findings by examining the CD4+ T-cell response to the major horse allergen Equ c 1, an important lipocalin allergen[8] with the prevalence of IgE reactivity close to 80% among horse

dust-allergic subjects.[9, 10] For this purpose, we analysed the CD4+ T-cell responses of horse dust-exposed Equ c 1-sensitized and healthy subjects focusing on the dominant epitope region of the allergen. This region is strongly recognized by the T cells of almost all Equ c 1-sensitized subjects examined.[11] As with the major allergen LY294002 in vitro of dog, Can f 1[1], and the major allergen of cow, Bos d 2[2], the frequency of Equ c 1-specific CD4+ T cells in the peripheral blood is very low. In allergic subjects, it is mostly higher than in non-allergic ones. Moreover, the function and phenotype of Equ c 1-specific CD4+ T cells differ between these two subject groups. p143–160 (GIVKENIIDLTKIDRCFQ), an 18-mer peptide containing the immunodominant

epitope Poziotinib in vivo region of Equ c 1, was synthesized and purified by GL Biochem (Shanghai, China). Recombinant (r) Equ c 1 was produced in Pichia pastoris, as described previously.[11] Fourteen clinically diagnosed horse-allergic subjects (subjects A–N) with positive (≥ 3 mm) skin prick tests with rEqu c 1 and nine horse dust-exposed non-atopic control Janus kinase (JAK) subjects (subjects O–W) with negative skin prick tests were recruited to the study. The subjects were characterized

at the Pulmonary Clinic of Kuopio University Hospital, as described in detail previously.[11] In brief, the allergic subjects exhibited a positive horse UniCAP result (FEIA; Pharmacia, Uppsala, Sweden; > 0·7 kU/l) and a positive skin prick test (≥ 3 mm) with a commercial horse epithelial extract (ALK Abellò, Hørsholm, Denmark), whereas the control subjects were negative in these tests. The non-atopic control subjects had horse riding as a hobby, and were therefore constantly exposed to horse allergens. Human leucocyte antigen (HLA) class II genotyping for the DQ and DR alleles of the subjects was performed in the Clinical Laboratory of the Finnish Red Cross Blood Service (Helsinki, Finland[12]) or in the Immunogenetics Laboratory of the University of Turku (Turku, Finland[13]) with PCR-based lanthanide-labelled sequence-specific oligonucleotide hybridization (Supplementary material, Table S1). Signed informed consent was provided by all subjects participating in the study and the study was approved by the Ethics Committee of Kuopio University Hospital, permission # 182/99.

His shirt was unbuttoned, his jacket discarded on the floor and,

His shirt was unbuttoned, his jacket discarded on the floor and, as I steered a wide berth around him, alarm bells started ringing in my head when he set his attention to unbuckling the belt of his trousers. Such a scene is not customary in the corridors of the Parasitology department of the University

of Heidelberg, not least at 9 am on a Monday morning, and to state that this spectacle caused quite a stir would be no understatement. Puzzled and perturbed, I nonetheless continued my walk to the lab, where I work on the Plasmodium parasite. I shall return to the topic of this gentleman because, although this tale doesn’t seem relevant for readers of an immunology journal, all will become clear later. The entire Parasitology department

in Heidelberg focuses on Plasmodium, and my particular interest is the immunology of this devious protist. Its complex and multifaceted life cycle starts with an infected mosquito see more (Figs. 1) taking a blood meal from an unfortunate mammal (Figs. 2), by which process parasites are deposited in the skin. A whistle-stop tour of the body then commences, as parasites travel from skin to blood, Erastin blood to liver, liver to bloodstream, with a quick pitstop at the lung before heading back to the bloodstream – only to be taken up by a mosquito again. The capacity of Plasmodium to metamorphose so dramatically and hijack so many components of the host organism has required the development of a neat box of tricks utilized to perplex the host immune response. Take, for example, antigenic variation. Infected erythrocytes adhere to host endothelial cells in the brain, liver, heart and

placenta. It is this sequestration, particularly in the brain microvasculature and placenta, which respectively leads to cerebral and pregnancy-associated malaria symptoms. Sixty var genes encode PfEMP1, (Plasmodium falciparum erythrocyte membrane protein 1) in ring stage parasitized erythrocytes, and PfEMP1 is known to bind CD36, ICAM-1 and other host receptors, mediating adhesion and promoting cerebral symptoms. The immune system diligently produces antibodies that inhibit binding of infected during erythrocytes and is capable of doing it quite well 1, but the parasite counters this by switching expression to another of its 60 var genes, and legions of new clones can surge forward uninhibited. Astoundingly, one of the var genes, var2csa, binds exclusively to the placenta and is only found in pregnant women, under the control of unique regulatory transcription mechanisms 2 capable of selective expression in pregnant hosts. One can only reluctantly admire the cunningness of this bug. The capacity for Plasmodium to confound the immune system also extends to T cells. Two classic proteins that have effectively become folklore in malaria circles are MSP-1 and CSP, the merozoite surface and circumsporozoite proteins, expressed in the blood and sporozoite stages respectively.

5b): 36% of activated Treg cells expressed SD-4, with more Treg c

5b): 36% of activated Treg cells expressed SD-4, with more Treg cells (53%) expressing JQ1 PD-1. Finally, we assayed the ability of SD-4+/+ versus SD-4−/− Treg cells to suppress T-cell activation (Fig. 6). Varying numbers of CD4+ CD25+ Treg cells purified from spleens of naive WT or KO mice were co-cultured with CFSE-labelled CD4+ CD25neg Tconv cells in the presence of anti-CD3 antibody and irradiated APC. T-cell proliferation was assayed by CFSE dilution. Without Treg cells, 60% of Tconv cells proliferated. As expected, SD-4+/+ Treg cells inhibited

this proliferation in a dose-dependent manner (down to 13% proliferation), and SD-4−/− Treg cells exhibited similar inhibitory capacity at every dose tested. These results show that SD-4 deficiency has little or no influence on Treg-cell function, thereby supporting the idea that exacerbation of GVHD by infusion of SD-4−/− T cells is primarily the result of augmented reactivity of Tconv cells to APC co-stimulation. SD-4 belongs to the SD family of transmembrane receptors heavily laden with heparan sulphate chains consisting of alternating disaccharide residues.[25] Because these heparan sulphate chains bind to a variety of proteins, including growth factors, cytokines, chemokines and extracellular matrices,[26] SD-4 can participate in a wide range of physiological and pathological

conditions. Indeed, SD-4 is known to play important roles in cell matrix-mediated and growth factor-mediated signalling

NVP-AUY922 molecular weight events.[27] SD-4-deficient mice may appear normal, but respond to intentional wounding with delayed repair, impaired angiogenesis, and poor focal adhesion of cells to matrix.[28] SD-4 also regulates immune responses: when given endotoxin, SD-4 KO mice succumb more readily to shock than WT controls;[29] SD-4 on B cells triggers formation of dendritic processes, which facilitate these cells’ interaction with other immune cells.[30] Our studies constitute the first evidence showing SD-4 on T cells to regulate the activation of allo-reactive T cells in GVHD. All the results using SD-4 KO mice unambiguously indicate SD-4 on T cells to be the sole DC-HIL ligand responsible for mediating its T-cell-inhibitory function (SD-4−/− T cells did not /www.selleck.co.jp/products/Fasudil-HCl(HA-1077).html bind DC-HIL nor did they react to DC-HIL’s inhibitory function), with one exception: DC-HIL-Fc treatment up-regulated cytokine production by SD-4−/− CD4+ T cells (compared with SD-4+/+ CD4+ T cells) following in vitro anti-CD3 stimulation (Fig. 2e). Because DC-HIL binds not only to a peptide sequence of SD-4 but also to saccharide (probably heparan sulphate or other structurally related saccharides),[6, 12] we speculate that absence of SD-4 and APC may restrict DC-HIL interaction exclusively to saccharides on T cells, thereby producing effects independent of SD-4.

The supernatant was used directly after clarification in some exp

The supernatant was used directly after clarification in some experiments, or in some cases, the fusion proteins were purified via the 6 × Histidine tag using Nickel-NTA agarose beads (Qiagen, Valencia, CA) and Poly-Prep® Chromatography

columns (BioRad, Hercules, CA) using the manufacturer’s recommendations. Interleukin-2 or the IL-2Rα chain was detected using either the anti-IL-2 monoclonal antibody (JES6-1A12; BD Pharmingen) or the anti-mouse IL-2Rα monoclonal antibody (PC61; BD Pharmingen), respectively. Wells of a 96-well plate were coated with either antibody (2·5 μg/ml) in PBS. Wells were blocked with 5% non-fat milk in PBS with 0·2% Tween (PBS-M-Tw) and fusion proteins were added for 1–2 hr at 37°. After PF-02341066 mouse washing, an anti-mouse IL-2 Epigenetics inhibitor biotin-labelled antibody (JES5H4; BD Pharmingen) was added and binding was detected using Strepavidin HRP (Southern Biotechnology Associates, Birmingham, AL). The ELISA plate was developed by adding 50 μl o-phenylenediamine (Sigma-Aldrich) in 0·1 m citrate buffer pH

4·5 and 0·04% H2O2, stopped by adding 50 μl/well 2 M H2SO4 and the absorbance was read at 490 nm. Immunoblot analyses were performed as reported previously with minor modifications.27 The following monoclonal antibodies were used: rat anti-mouse IL-2 antibody (JES6-1A12; BD Pharmingen), rat anti-mouse IL-2Rα (PC61; BD Pharmingen), and mouse anti-6 × His monoclonal antibody (MM5-156P; Covance, Princeton, NJ). Detection was performed using a goat anti-rat

HRP-conjugated antibody (Jackson Immuno Research, West Grove, PA) and developed using the Amersham ECL Plus Western blotting detection reagent (GE Healthcare) following the manufacturer’s recommendations. A determination of fusion protein concentration new was established using immunoblot analyses and quantitative densitometry and compared with recombinant IL-2. For MMP immunoblot analyses, extracts or supernatants were probed with goat anti-mouse MMP2 or MMP9 antibodies (R&D Systems, Minneapolis, MN). Fusion proteins were digested with PSA (Cortex Biochem, San Leandro, CA) or prostate extracts in 50 mm Tris–HCl, 100 mm NaCl pH 7·8 at 37°. For digestion of the fusion protein containing the MMP cleavage sequence, MMP9 or MMP2 (R&D Systems) was activated with p-aminophenylmercuric acetate and this activated protease or equivalent amount of activating solution without the protease was used to digest the fusion protein for 1 hr at 37° for MMP9 and 10 min for MMP2. Aliquots of digests were loaded on 15% Laemmli gels for Western blotting.

The use of anthelmintics for the definitive hosts is difficult in

The use of anthelmintics for the definitive hosts is difficult in most third world countries, and alternative strategies are needed. Interruption of the hydatid life cycle within the intermediate host by vaccination against the larval stage may be a viable supplement to anthelmintics (2,3,6,7). In the 1960s, it was discovered that the secreted proteins of the oncosphere induce protection. EG95 was subsequently identified as a protective antigen

when immunized animals were challenged with E. granulosus eggs (8). In addition, the antibody produced by animals vaccinated with E. granulosus oncospheres or the EG95 protein was shown to be highly effective in a complement-dependent in vitro oncosphere-killing assay (6,9,10). Poxviruses offer an KU-60019 concentration efficient, low-cost means by which foreign antigen can be delivered to target species (11). Recombinant vaccinia virus (VACV) has been successfully

Decitabine concentration used to vaccinate against rabies in Europe and in America (12,13). In this study, we explored the use of VACV as a viral delivery vehicle for the hydatid oncosphere antigen EG95 in a mouse model and in sheep. We show that antiserum produced in mice against the EG95 antigen is effective in killing E. granulosus oncospheres in an in vitro assay. The coding region of the E. granulosus protective antigen EG95 (7,8) was inserted at the thymidine kinase gene of the VACV Lister strain (termed VV399). The construction of VV399 is described in (14,15). Immunization of mice with VV399: Balb/C mice 6–8 weeks of age were anaesthetized with approximately 200 μL avertin [2,2,2, tribromoethanol; 0·2 mL/15 g mouse of 20 mg/mL solution (Sigma-Aldrich, St. Louis, MO, USA)] injected intraperitoneally. Mice were infected intranasally with 50 μL containing 1 × 108 pfu of VV399. Twenty-five microlitre was introduced into each nostril

using a syringe. Intraperitoneal immunization with EG95 protein: Balb/C mice were immunized with 10 μg of EG95-6xHIS (cloning and expression described in 16) in a total volume 250 μL via the Cytidine deaminase intraperitoneal route. Alum adjuvant was prepared as described by Herbert (17). Antigen was prepared by mixing equal parts of soluble protein antigen with adjuvant. Groups of mice were held in individual isolator cages during the course of the experiment. Mice were weighed every 2 weeks following primary immunization and booster immunization. Outbred sheep of mixed sex and <1 year of age were first tested for antibodies against EG95 antigen by ELISA. Animals were divided into two random groups. Group 1: Six sheep were immunized by scarification with 108 pfu of VV399 in PBS in a total volume of 100 μL. A 4 × 4 cm scratched area was made on the bare skin on the inside of each back leg, and 50 μL of virus applied. Group 2: Six animals were each immunized with 50 μg GST-EG95 protein (cloning and expression described in 7,8) with 1 mg QuilA.

9 and 7 0 pg mL−1, respectively Secretions of IFN-γ and IL-10 in

9 and 7.0 pg mL−1, respectively. Secretions of IFN-γ and IL-10 in response to a given antigen were considered positive when absolute concentrations were ≥100 and ≥29 pg mL−1, respectively, and E/C was ≥2 (Brock et al., 2004; Moura et al., 2004; Al-Attiyah & Mustafa, 2008). A positive response for both cytokines was considered strong

at ≥60%, moderate at 40% to <60% and weak at <40% (Mustafa, 2009a, b). The ratios of IFN-γ : IL-10 were calculated to determine Th1 vs. anti-inflammatory biases in response to Con A, complex mycobacterial antigens and peptides of RD1 and RD15. The ratios of ≥2 were considered to be Th1, <0.5 to be anti-inflammatory and 0.5 to <2 to be neither Th1 nor anti-inflammatory. Moreover, Th1 responses were considered strong, moderate and weak with IFN-γ : IL-10 ratios of >20, 5–20 and 2 to <5, respectively. The antigen-induced cell proliferation and IFN-γ secretion results LEE011 supplier with Con A, complex

mycobacterial antigens and peptide pools were statistically analyzed for significant differences between TB patients and healthy subjects using the nonparametric Mann–Whitney U-test for two independent samples. P-values of <0.05 were considered significant. In lymphocyte proliferation assays, Con A and the complex mycobacterial antigens were strong stimulators of PBMC from TB patients and healthy subjects, as indicated by high percentages of positive responders (83–100%) (Fig. 1a and www.selleckchem.com/products/pifithrin-alpha.html b). Furthermore, the proliferation of PBMC

from TB patients was strong in response to RD1 peptide pool (70% positive responders) and weak in response to peptide pools of RD15 and all of its ORFs (<40% positive responders) (Fig. 1c). In healthy subjects, the RD1 peptide pool induced moderate responses (47% positive responders), whereas the peptide pool of RD15 and 1502 induced strong responses (70% and 63% positive responders, respectively), and RD1501, RD1504 and RD1505 induce moderate responses (40%, 43% and 43% positive responders, respectively) (Fig. 1d). Peptide pools of other ORFs of RD15 induced weak proliferation of PBMC (<40% positive responders) (Fig. 1d). Statistical analysis of the results showed that positive responses induced by RD15 and RD1502 were significantly higher (P<0.05) in healthy Masitinib (AB1010) subjects than in TB patients (Fig. 1c and d). To further determine the secretion of Th1 and anti-inflammatory cytokines and their ratios in response to complex mycobacterial antigens and peptides of RD1 and RD15, we studied secretion of Th1 cytokine IFN-γ and the anti-inflammatory cytokine IL-10 with PBMC from 20 TB patients and 12 healthy subjects using FlowCytomix assays. The results showed that PBMC from both TB patients and healthy subjects secreted high concentrations of IFN-γ (median values=6727–10 986 pg mL−1) with strong responses to complex mycobacterial antigens (positive responders =92–100%) (Fig. 2a and b).

Lung cells

were also stained for the following combinatio

Lung cells

were also stained for the following combinations; CCR3+ MBP+, IL-5Rα+ CCR3+ and IL-5Rα+ MBP+ cells. Cells were pre-treated with 2% mouse serum (DAKO, Carpinteria, CA) for 15 min to prevent non-specific binding and thereafter stained with antibody or the appropriate isotype control antibody in saturating concentrations. The cells were incubated for 30 min at 4° with antibodies or isotype control, followed by two washing steps. Finally, the samples were fixed in 2% paraformaldehyde and kept at 4° until flow cytometric analysis. In experiments where cells were stained for surface marker and intracellular stained for MBP, an extended protocol was used, as per the manufacturer’s instructions (BD Cytofix/Cytoperm™ Fixation/Permeabilization Solution buy Z-VAD-FMK kit; Cat no.: 554722). In some experiments, cells were stained with 7-aminoactinomycin D (7-AAD) to exclude dead cells. In other studies, cells were also stained with anti-CD45 PerCP to exclude non leucocyte cells. The flow cytometric analysis was carried out using a FACScan flow cytometer (BD Bioscience). Twenty thousand cells were computed in list mode and Maraviroc analysed using the cellquest pro software. Gating was set on all intact cells and

cells with the CCR3+ high side scatter (SSChigh) profile were identified as eosinophils. As eosinophil-lineage-committed progenitors are found in the mononuclear cell population,24 gating was also made Clomifene on cells with an SSClow profile (Fig. 1b). Animals were sensitized and exposed to OVA and lung and BM cells were harvested as described above for in vitro lung and BM colony assay. Lung CD34+ progenitor cells were enriched from the sampled Percoll fractions as described above. Enrichment of BM CD34+ cells was performed as previously described with some modification.9

Briefly, mononuclear CD3+ cells and neutrophils were depleted using biotinylated antibodies and finally CD34+ cells were enriched using the same magnetic separation method as above. Both BM and lung CD34+ cells (5 × 105) were cultured at 37° in 5% CO2 in a 12-well plate in 1 ml RPMI-1640 culture medium completed with 0·9% methylcellulose, 20% FCS, 1% penicillin-streptomycin, 2 mm l-glutamine and 0·0006%β-mercaptoethanol (all obtained from Sigma-Aldrich). Cells were seeded and divided into groups depending on cytokines added: control (no cytokines added), recombinant murine IL-5 (rmIL-5; 10 ng/ml; R&D Systems), rmEotaxin-2 (500 ng/ml; PeproTech EC, London, UK) and rmIL-5 together with rmEotaxin-2 (10 and 500 ng/ml, respectively). The BM and lung cultures were fed with 100 μl RPMI-1640 completed with penicillin-streptomycin, l-glutamine and the respective cytokines on day 6 of culture. The BM colonies were counted on day 8 of culture and lung colonies were counted on days 8–14 of culture, using an inverted light microscope as described previously.25 Animals were sensitized and exposed to OVA or PBS and BrdU was administered as described above.