The neutralizing mAb mixture prevented acquisition whereas the no

The neutralizing mAb mixture prevented acquisition whereas the non-neutralizing mAb mixture did not. On the other hand, this mixture afforded post-infection control of viraemia, suggesting that Fc-mediated effector function contributes to this type of protection. Similar results were reported for another antibody specific for the immunodominant region of gp41 but no functional

Pembrolizumab molecular weight data other than virus capture was provided in that study.[16] Post-infection control is also a common finding for neutralizing mAbs used at doses insufficient to block acquisition (summarized in ref. [19]). Given that the in vivo half-lives of mAbs are short, typically ranging from 3 days to 2 weeks, they must exert their activities early after passive immunization as post-infection control by Fiebig Stage VI.[19] The short-term effect probably is to protect components of the immune system early in infection such that they can mature and mediate post-infection control after mAb decay. This possibility is supported by studies in mice showing that NK-mediated lysis of target cells expressing a foreign antigen early in the immune response results in strong CD4+ T Palbociclib cell, CD8+ T cell and antibody responses downstream to release of the foreign antigen.[73] It is reasonable to expect that a similar

phenomenon would follow ADCC-induced lysis of target cells early in infection. This form of Fc-mediated protection would be most important in limiting the expansion of the local PAK5 founder population or perhaps decreasing systemic viral spread (Fig. 3). Correlations have been reported repeatedly between ADCC or ADCVI and post-infection control in vaccinated NHPs,[74-78] supporting this possibility. Despite the repeated correlations between Fc-mediated effector function and post-infection control in both active and passive immunization studies in NHPs, no study shows that passive immunization with a non-neutralizing mAb can block acquisition. Until a definitive passive immunization study employing a non-neutralizing antibody with Fc-mediated effector function, including an attenuated LALA variant as a negative

control, either rules this possibility in or out, the field is left with correlations. Two recent NHP vaccine studies report an inverse correlation between reduced acquisition and ADCC titres.[79, 80] In addition to the NHP studies, increasingly solid support indicating a role of Fc-mediated protection in preventing acquisition is developing from studies of infected and vaccinated humans. A recent study in HIV-infected mothers with high viral loads showed an inverse correlation between ADCC titres in breast milk and probability of transmission to their infants.[81] No such correlation was found for neutralization.[81] The earliest vaccine study reported an inverse correlation between ADCVI titres and risk of infection in a subgroup of vaccines in the VAX004 Phase III efficacy trial, although no overall protection was observed.

Recently several methods, especially methods based on flow cytome

Recently several methods, especially methods based on flow cytometry, have emerged, avoiding the use of radioactive isotopes. Several fluorochromes that can be integrated into the target cells have been used

in a manner similar to 51Cr [2, 3]. However, the spontaneous release of these fluorescent dyes can U0126 chemical structure also be high, with possible labelling of other cells, thus preventing sufficient discrimination between target and effector populations [4]. In this study we present adaptions to an assay, described thoroughly by Bryceson et al. [5], by flow cytometric assessment of CD107a surface expression. This assay detects the amount of possible effector cell degranulation 3 Methyladenine in response to recognition of antibodies bound to epitopes presented on the target cells, rather than measuring target cell lysis directly. Upon stimulation with appropriate target cells, the effector cells will release the assayed cytotoxic proteins by fusion of secretory lysosomes with the plasma membrane,

thereby effecting target cell lysis [6]. This type of assay is used increasingly for measuring NK cell cytotoxicity [7], but it is also applicable for other types of cytotoxic effector mechanisms. With the present optimized assay we analysed different aspects of cytotoxicity reactions and the potential consequences of HERV epitope expression on MS patient PBMCs. Polyclonal antibodies against learn more defined peptides, derived from specific sequences in the Env- and Gag-regions from HERV-H/F and HERV-W, were raised in rabbits. By including or excluding these antibodies in the test it is possible to assess the action of both antibody-dependent and -independent cytotoxic cell populations towards cells expressing these viral peptides/epitopes. Thus, the test contributes information about both the relevance of the constructed peptides/epitopes and also the pathogenic potential of these, when ‘seen’ by the cytotoxic cell populations. The results

then lead to subsequent analysis of both the level of cytotoxic antibodies in MS patients and to the testing of possible pathogenic activation of cytotoxic cells in the patients, thereby gauging the potential of own lymphocytes in reactions against ‘self’ or ‘self with up-regulated HERV expression’. For the present study, PBMCs from 10 healthy donors [five females (aged 24–52 years), five males (aged 27–62 years)] were used as effector cells in NK and ADCC assays. Venous blood was drawn and processed on the same day in our laboratory or the respective clinics. PBMCs were prepared by standard Isopaque-Ficoll centrifugation. The separated cells were aliquoted and cryopreserved in RPMI-1640 with the addition of 20% human serum (HS) and 10% dimethylsulphoxide (DMSO) at −135°C until use.

Similarly, 2 × 106 CD19+ B cells were added to equal numbers of i

Similarly, 2 × 106 CD19+ B cells were added to equal numbers of iDC in the presence or absence PD98059 datasheet of the pan-RAR selective antagonist ER50891 (Tocris Biosciences, Minneapolis, MN, USA) at a final concentration of 1 μM for 72 h. The B cells and/or DC were subsequently isolated by magnet assistance for further analysis. Statistically relevant differences among means (Student’s t-test, analysis of variance: anova) and medians (paired Wilcoxon’s test) were ascertained using GraphPad Prism version 4 software (GraphPad, La Jolla, CA, USA). In all statistical analyses, a P-value < 0·05 was considered to represent

statistically significant differences. We have shown previously that T1D patients treated with cDC or iDC exhibit an increase in the frequency of B220+CD11c– cells in the peripheral blood [31]. Flow cytometry of these cells [31] suggested that they represented a late transitional B cell population that shared some cell surface proteins (CD5+CD10+CD24+CD38intermediate) with at least one population of human Bregs recently reported and characterized [23, 32, 33]). Thus, we hypothesized that the increase in the frequency of B220+CD11c– cells in DC recipients was

a consequence PI3K Inhibitor Library molecular weight of, and reflected an increase in, the number of constituent suppressive immunoregulatory B cell populations that express B220 on the surface, even though B220 on its own does not define B cells [29, 30]. We discovered subsequently that a population of CD19+B220+CD11c– IL-10+ cells accounted for an average of 48% of the B220+CD11c– cells (V. D. C., B. P. and N. G., unpublished data) and, more importantly, that the CD19+B220+CD11c– IL-10+ population was immunosuppressive in PTK6 vitro [31]. To date, two human B cell populations with immunosuppressive ability in vitro have been characterized, mainly by cell

surface markers [23, 25, 26, 32, 40]. Although both populations produce IL-10, their surface phenotypes are different. ‘B10’ Bregs express the CD1d and CD5 markers [25, 26], whereas the other suppressive cells are characterized specifically as CD19+CD24+/intermediateCD38+/intermediate [23, 32, 40]. We first asked if the suppressive properties of the CD19+B220+CD11c– IL-10+ B cells shown in [31] were concentrated in either or both of the currently characterized Bregs (CD19+CD1d+CD5+ or CD19+CD24+CD27+CD38+ B cells [23, 25, 26, 32, 40]), or if other novel CD19+ cell populations inside the parental CD19+B220+CD11c– IL-10+ cell population possessed suppressive ability. Using flow cytometry (Supplementary Fig. S1 shows the approach), we determined that CD19+CD24+CD27+CD38+ cells accounted for 19·85% (median) of FACS-sorted CD11c–B220+CD19+ IL-10+ cells from freshly acquired PBMC (Fig. 1a; n = 6 healthy unrelated adult individuals). We did not detect any B10 Bregs (CD19+CD1d+CD5+ IL-10+ cells) [25] inside the CD11c–B220+CD19+ IL-10+ population (not shown).

Clearly, as low vitamin D status and its clinical consequences ma

Clearly, as low vitamin D status and its clinical consequences may be secondary to a host of factors, including advanced age, reduced mobility from disease, reverse causation cannot be excluded. Studies investigating the effect of migration and vitamin D supplementation on PD risk are lacking. There is a clear heritable component in PD. Genetic studies have pointed to a possible role of vitamin D in susceptibility to the disease. Polymorphisms in the VDR gene have been shown to associate with PD risk

in American and Korean cohorts, with the former cohort also showing an age of onset effect [138, 139]. The relatively small sample sizes and the inconsistent replication of SNPs in the VDR gene in discovery and validation sets dampen the impact of these findings. GWAS have identified an increasing number of candidate Olaparib solubility dmso risk genes in PD, several of which have VDR-binding sites closely associated with them raising the possibility that vitamin D may influence their expression. The biological relevance of a subset of these

susceptibility genes with associated VDR binding on brain function has been well delineated with evidence for roles in nigrostriatal dopaminergic neurotransmission, neurogenesis and neurite outgrowth, and neural ectodermal expression (especially within the marginal and subventricular zones) (see Table 2) [140-144]. Amyotrophic lateral sclerosis (ALS) is a progressive U0126 mouse neurodegenerative disease affecting both the central and peripheral nervous systems [145]. ALS pathology reveals degeneration of motor neurones and corticospinal tracts, brainstem nuclei, and spinal cord anterior horn cells, with a subset of patients having intracytoplasmic transactive responsive DNA-binding protein inclusions (TDP-43) [146]. Multiple effector pathways are thought to contribute to ALS pathology including neurotrophic factor deficiency, glutamate toxicity, and damage from ROS [54]. Given that many of these effector

pathways are influenced by vitamin D in rodent models, there has been growing interest in the concept that this secosteroid may influence susceptibility to and disease progression in ALS. The epidemiological evidence incriminating vitamin D as a possible risk factor in ALS is sparse. The relatively Phosphoprotein phosphatase low population prevalence probably contributes but there may be no association. Season of birth observations have been conflicting with a few studies reporting excess births between April and July [147], and others reporting birth excess in between October and December (with a trough between April and July) [148]. A latitude gradient has been suggested, but the results are divergent. An American cohort outlining the geographic distribution of ALS using mortality data demonstrated a north-west to south-east gradient [149], a finding mirrored in a more recent study which found a higher ALS-associated death rate in more northern states [150].

4a,b; NS=42·77 (33·80–64·12) versus ML = 94·09 (46·72–97·90); P <

In addition, cell frequency also increased in the ML-stimulated PBMC culture of RR/HIV patients

when compared with the HC and RR groups under the same conditions [Fig. 4a,b; HC = 15·35 (0·5–28·08), RR = 9·87 (4·50–38·08); P < 0·05]. The frequency of CD4+ CD25+/CD4+ T cells and CD8+ CD25+/CD8+ T cells mTOR inhibitor was not significantly modulated in any of these groups (data not shown). As leprosy is marked by a localized immune inflammation in skin lesions, the expression of these activation markers in the skin biopsies of the RR and RR/HIV patients was evaluated. Double-immune labelling was used to examine CD69 and CD38 activation markers in CD4+ and CD8+ T cells in RR and RR/HIV skin lesions. Both groups presented a dermal infiltrate consisting of numerous CD3+ CD4+ and CD3+ CD8+ T cells (data not shown). The percentage of CD4+ CD69+ cells found was similar in both the RR (50%) and RR/HIV (40–50%) lesions (Fig. 3c). In contrast, a greater percentage of

CD4+ T cells co-localizing with CD38 (40–50%) was observed among the RR/HIV patients. This pattern differed from the one seen in RR lesions in which only a few cells co-localized with CD38 (< 5%). RR/HIV dermal infiltrate also presented greater numbers of CD8+ CD69+ T cells than those found among the RR patients (Fig. 4c; RR 20% versus RR/HIV 50%), and of CD8+ CD38+ T cells (Fig. 4c; RR< 5% versus RR/HIV40–50%). Memory T cells are known to be more XAV-939 nmr sensitive to antigenic stimuli than naive T cells and to mount a more rapid and broader pathogen-specific response.[25] As antiretroviral therapy leads to an increase in memory T cells[26] and all patients evaluated in this study were under HAART treatment, the next step was to evaluate the memory phenotype of the PBMCs of RR/HIV patients after ML in vitro stimulation via analysis of molecular surface expression of CD45RA and CCR7. In compliance with these parameters, T selleck cells were classified as naive T cells (CCR7+ CD45RA+), central memory T cells (TCM; CCR7+ CD45RA−), effector memory T cells (TEM; CCR7− CD45RA−),

or TEMRA cells (CCR7– CD45RA+).[27] In ML-stimulated cultures, an increase in TCM CD4+ T-cell frequencies was observed in both the RR and RR/HIV groups [Fig. 5a,b; RR NS = 16·5 (10·2–23·20) versus ML = 22·5 (19·5–30·3); P < 0·05; RR/HIV NS = 10·8 (9·8–20·9) versus ML = 23·8 (16·15–36·1)]. The same profile was identified in relation to TCM CD8+ cell frequencies in the RR/HIV group alone [Fig. 5a–c; NS = 11·7 (7·8–18·9) versus ML = 20·40 (10·5–28·4); P < 0·05]. In this group, an increase in TEM CD8+ T cells was also seen in ML-stimulated cells in comparison to NS cells [Fig. 5a–c; NS = 16·4 (7·4–23·7) versus ML = 27·50 (22·3–43·3); P < 0·05] and also in comparison with ML-stimulated cells of the other groups evaluated [Fig. 5a–c; HC 10·88 (9·2–22·10); RR 15·17 (4·3–24·6); RR/HIV 27·4 (22·3–43·3); P < 0·05].

The increase in larvae from day 3 to 7 days post-challenge was pr

The increase in larvae from day 3 to 7 days post-challenge was probably due to the gradual migration of L3 from the stomach to the different sections of the small intestine (24,25). Individuals never completely cleared the infection, and nematodes were still present, although with very low numbers, in the first section at 120 days post-challenge. Graphidium strigosum: Abundance was consistently higher in the fundus compared to the antrum, and no temporal changes were DAPT in vivo observed between sampling points (or the interaction between sampling point and

organ section), when differences among individuals and the nonindependent sampling of the two parts of the stomach from the same individual were considered (Figure 1b, Table 2). All infected individuals maintained a constant number of nematodes up to 120 days post-infection. The drop in parasite number selleck kinase inhibitor in the antrum at day 40 and 60 post-challenge was caused by a sampling procedure and should not be considered biologically relevant. These

results were consistent with our long-term observations on the intensity of infection of these nematodes in free-living rabbits of different age, specifically, rabbits can reduce or clear T. retortaeformis but not G. strigosum. Trichostrongylus retortaeformis: A strong IFN-γ expression in the first section of the small intestine of infected rabbits was observed during the first 30 days post-challenge; thereafter, no dominant pattern was observed (Figure 2a). Analysis based on the normalized Ct values (that differs from the 2−ΔΔCt transformation in Figure 2) found that changes in IFN-γ and IL-4 significantly Flavopiridol (Alvocidib) differed between treatments (infected and controls) and time post-infection (DPI): IFN-γ decreased while IL-4 increased in transcription with the infection course, IL-10 exhibited constant expression over time although was significantly higher in infected compared to controls (Table 3). Graphidium strigosum: A robust IL-4 expression was observed in the top section of

the stomach of infected rabbits; however, the between-individual variability was high as highlighted by the large standard error bars (Figure 2b). Based on the Ct values, the expression of the three cytokines was higher in the infected compared to the controls but no significant changes were recorded during the course of the infection (Table 3). The two infections clearly showed different cytokine profiles, which imply differences in the effectors and timing of their activation as well as their dynamical consequences. The somatic antibody response of infected rabbits to L3 and adult stage was similar both for IgA and IgG against the two nematodes supporting the hypothesis that the two parasite stages cross-react at the antibody level. As such, we only present the results for the adult stage (Figures 3 and 4) and summarize in the supplement the findings for the L3 stage (Figures S1 and S2).

The differences in the complexity of the CD8+ T-cell response or

The differences in the complexity of the CD8+ T-cell response or the influence

of background genes (e.g. extent of IFN-γ production) may account for the results. Using LCMV infection of naïve C57BL-6-PKO mice Lykens et al. recently showed that heightened antigenic stimulation is responsible for exaggerated T-cell activation [[49]]. They suggested that perforin-dependent cytotoxicity, in addition to promoting viral clearance, regulates T-cell activation by modulating Ag presentation [[49]]. Despite the differences in susceptibility of naïve BALB/c and C57BL/6 PKO mice to LCMV infection, we also observed massive CD8+ T-cell expansion and accelerated LCMV-induced mortality in GP33-vaccinated compared with naïve C57BL/6-PKO selleck screening library mice (data not shown). Thus, the vaccine-induced sensitization to mortality associated with PKO memory CD8+ T cells after LCMV infection is not restricted to BALB/c background. In addition, functional exhaustion of antigen-specific CD8+ T cells is not always associated with chronic infection [[50, 51]]. Chronic infection may be pathogen or host specific and it does not necessarily lead to Ag-specific CD8+ T-cell

exhaustion in all the cases. Although we observed lesser degree of “exhaustion” as characterized by TNF and PD-1 expression in GP283-specific CD8+ T cells compared with NP118-specific CD8+ T cells, viral control was not achieved in the absence of perforin in both cases (Fig. 5). In the absence of perforin, the phenotype of GP283-specific CD8+ T cells appeared “less exhausted” at the time we analyzed them could reflect the click here extent that these cells can regulate cytokine production. In addition, it remained to be elucidated whether encounter with antigen is similar between the NP118- and GP283-specific memory CD8+ T

cells in the PKO mice, not just initially, but throughout the infection course. Previous studies using different models of infection showed that protective immunity mediated by pathogen-specific CD8+ T cells did not correlate with immunodominance hierarchies after infection [[36, 37]]. Based on the results with PKO mice vaccinated with dominant NP118 epitope, we expected that massive antigen-specific memory CD8+ T-cell expansion contributed to the LCMV-induced mortality independent of epitope specificity. Interestingly, learn more PKO mice vaccinated with subdominant GP283 epitope survived the LCMV infection even though they contained similar starting memory CD8+ T-cell numbers and underwent similar expansion in numbers as NP118-specfic CD8+ T cells. These results suggested that epitope specificity dictates the LCMV-induced mortality in vaccinated PKO mice. Furthermore, we also observed less cytokine dysregulation, in particular IFN-γ, by GP283-specific CD8+ T cells following LCMV infection. It is unclear which specific parameter(s) influence the cytokine profile of these GP283-specific CD8+ T cells and subsequent vaccine-induced mortality in PKO mice.

The RNA concentration and purity was measured by a spectrophotome

The RNA concentration and purity was measured by a spectrophotometer (ND-1000; NanoDrop Technologies Inc.). Reverse transcription was performed with TaqMan reverse transcription reagents (Applied Biosystems). Quantitative PCR was performed using StepOnePlus instrumentation (Applied Selleck I BET 762 Biosystems) with TaqMan Fast Universal PCR Master Mix and predesigned FAM-labelled gene expression assay reagents (Applied Biosystems). Selected cytokines and transcription factors were IL-17A (cat. no. Hs00174383_m1), FoxP3 (Hs00203958_m1), RORc (cat. no. Hs01076112_m1) and IFN-γ (Hs00174143_m1). Ribosomal 18 s RNA served as the endogenous control (Hs99999901_s1). The quantities of target gene

expression were analysed by a comparative threshold cycle (Ct) method (as recommended by Applied Biosystems). An exogenous cDNA pool calibrator was collected from phytohaemagglutinin (PHA)-stimulated peripheral blood mononuclear cells (PBMC) and considered as an interassay standard to which normalized samples were compared. ΔCt stands for the difference between the Ct of the marker gene and Ct of the 18S gene, whereas ΔΔCt is the difference between the ΔCt of the sample and ΔCt of the calibrator. Calculation of 2−ΔΔCt then gives the relative amount of target gene in the sample compared with the calibrator, both normalized Pirfenidone to

an endogenous control (18S). For presentations the relative amounts (2−ΔΔCt) of target genes were multiplied by a factor 1000 and expressed as relative units. If the samples Ct value for target gene did not reach quantitative Nitroxoline level, then an artificial value that was half the lowest quantitative value in relative units was given to the sample. We cultured small intestinal biopsy samples from 23 patients with untreated CD (of which six also had T1D) and 10 reference children (five positive for TGA) for 72 h in RPMI-5% human AB serum and measured the concentration

of IL-17, Il-1β and IL-6 secreted in the culture supernatants by using flow-cytometric bead array (Bender Medsystems, Vienna, Austria). Samples below the detection limit (or the cut-off level) of the method were considered as undetectable, but were given half the cut-off value to enable statistical analyses. The human colon adenocarcinoma cell line (CaCo-2) was obtained from American Type Culture Collection (ATCC) (Teddington, UK). Cells were grown in Eagle’s minimal essential medium (Sigma) containing 10% heat-activated and sterile filtered fetal bovine serum (FBS) supplemented with penicillin (0·1 g/l) and streptomycin (0·15 g/l) at + 37°C and 5% CO2. CaCo-2 cells were grown in a 75 cm2 flask for 6 days and were thereafter plated into sterile 48-well plates (Greiner Bio-One GmbH, Frickenhausen, Germany) and grown for 4 days at a density of 1·5 × 105 cells per well and a final volume of 0·5 ml/well. The cells were incubated for 5 h with recombinant human IL (rhIL)-17 (1 or 50 pg/ml; cat. no.

(V ) braziliensis compared to those in the animals infected with

(V.) braziliensis compared to those in the animals infected with L. (L.) amazonensis. Interestingly, this change was just noted when experimental infections had opposite evolvements; while BALB/c mice infected with L. (L.) amazonensis developed a severe infection, with an increase in the lesion size, high tissue parasitism, and pathological process in the skin associated

with tissue destruction, the animals infected with L. (V.) braziliensis showed minimal skin lesions, scanty parasitism and slight Y 27632 pathological events in the skin sites of infection, thus suggesting that the early response of both DCs subsets in L. (L.) amazonensis BALB/C mice infection was unable to control the infection, despite a high expression of CD4+ cells. In contrast, the increase in these DCs subsets population was correlated with the regression of the L. (V.) braziliensis infection at 8th weeks PI and the increase in the number of CD4+ and CD8+ cells in the lesion site. These experimental differences in the immunopathogenic competences of parasites belonging to the subgenus Leishmania and Viannia seem to

confirm prior evidences looked at a clinical–immunopathological level of ACL because of L. (V.) braziliensis and L. (L.) amazonensis (5). Corroborating with the above results, it is worth noting that the experiment using DCs derived from human PBMC showed that L. (L.) amazonensis

was able to abrogate check details full DCs differentiation, decreasing the expression of co-stimulator molecules and cytokines production, and not only causing a delay in the immune response but also favouring the establishment of L. (L.) amazonensis in the human host (20). Another study showing that DCs derived from L. (L.) amazonensis-infected mice were less potent in activating the IL-12-producing CD11c DC subsets, thus preferentially activating CD4+ T cells with IFN-γlow IL-10high 4-Aminobutyrate aminotransferase phenotypes (21), should also be highlighted. In addition, DCs infected with the amastigote form of L. (L.) amazonensis were less mature and less potent antigen-presenting cells than those infected with promastigote, as jugged by the lower expression of co-stimulatory molecules, suppressed IL-12 and increased IL-10 expression under positive stimuli, and reduced effectiveness for priming CD4+ T cells from naïve and infected mice, suggesting that L. (L.) amazonensis, specially its intracellular form, has developed strategies to down-regulate early innate signalling events, resulting in impaired DCs function and Th1 inactivation (22). By the other site, DCs experimentally infected with the promastigote form of L. (V.) braziliensis up-regulated activation markers, leading to a production of IL-12 and TNF-α.

Recently, we have demonstrated that RBV down-modulates inducible

Recently, we have demonstrated that RBV down-modulates inducible co-stimulator (ICOS) on human CD4+ T cells, which in turn decreases IL-10 secretion, leading to the maintenance of Th1 activity,[30] and speculated that RBV might affect Treg cells that also express ICOS on their surface. In the present study, we examined the effects of RBV against human peripheral Treg cells in vitro and found the unique characteristics of RBV, which might down-modulate the activity of Treg cells by inhibiting the differentiation of naive CD4+ T cells into Tregadapt cells. Peripheral blood was obtained from five healthy individuals

who were serologically confirmed to be free from hepatitis B virus, HCV, or human immunodeficiency virus infection. This study protocol conformed to the ethical guidelines of the Declaration of Helsinki as reflected in a priori approval by

the Institutional Copanlisib Review Committee of Nippon Medical School. CD4+ T cells were purified from peripheral blood mononuclear cells (PBMCs) isolated from heparinized blood using the Ficoll–Paque (Amersham, Buckinghamshire, UK) Lumacaftor purchase density-gradient method with a magnetic cell sorter (Miltenyi Biotech, Auburn, CA). Briefly, PBMCs were incubated with a CD4+ T-cell isolation cocktail containing biotin-conjugated anti-human CD8, CD14, CD16, CD19, CD36, CD56, CD123, T-cell receptor-γδ, and glycophorin A antibodies 17-DMAG (Alvespimycin) HCl (Miltenyi Biotech) for 10 min at 4° and additionally labelled with magnetic bead-conjugated streptavidin for 15 min at 4°. Cells were washed, subjected to LS separation columns, and the pass-through fraction was collected as CD4+ T cells. Because Treg cells could be identified by their CD127 deficiency,[31] CD4+ T cells were subsequently

divided into CD25− and CD25+ CD127− cell fractions using FACSort. Briefly, CD4+ T cells were stained with FITC-conjugated anti-human CD25 (BD-Bioscience, San Diego, CA) and Alexa-Fluor647-conjugated anti-human CD127 monoclonal antibodies (mAbs) (BD Bioscience). Cells were sorted into FACS AriAll (BD Bioscience) and both CD25− and CD25+ CD127− cells were collected. All cells were cultured in complete T-cell medium, RPMI-1640 medium supplemented with 10% heat-inactivated fetal calf serum, HEPES-buffer solution 5 mm, penicillin 100 U/ml, streptomycin100 μg/ml, l-glutamine 2 mm, sodium pyruvate solution 2 mm, and non-essential amino acid solution 2 mm (all these supplements were purchased from Gibco-BRL, Santa Clara, CA), modified vitamins 2 mm (Dainippon Pharmaceutical Co. Ltd., Tokyo, Japan), and 2-mercaptoethanol 2 mm (Sigma Chemical Company, St Louis, MO). Anti-human IL-10 and anti-human transforming growth factor-β1 (TGF-β1) mAbs (e-Bioscience, San Diego, CA) were used for cytokine-neutralizing assays.