Taking into account the fact that LTC4 imposes changes in DCs tha

Taking into account the fact that LTC4 imposes changes in DCs that prevent their maturation we decided to evaluate their impact on the genesis of the LY2606368 in vitro adaptive response, through the analysis of the cytokines induced. With this aim, immature and activated DCs were cultured for 18 hr at 37° in presence or not of LTC4 (10–8 m). After incubation, culture supernatants were collected and we evaluated cytokines by ELISA. As shown in Fig. 3(a), LTC4 increased the production of TNF-α in immature DCs but was unable to reverse its release induced by LPS. Interestingly, LTC4 completely abolished the induction of IL-12p70 in LPS-stimulated DCs (Fig. 3b), indicating

an antagonistic effect of LPS. Therefore, LTC4 inhibits the induction of a Th1 profile by T CD4+ naive lymphocytes, by acting on activated DCs.34,35 Moreover, to further investigate the effect of LTC4 we decided to evaluate whether LTC4 could favour a tolerogenic state;36,37 however, when we analysed the release of IL-10 in culture supernatants, VX-765 we showed inhibition of this cytokine in LPS-treated DCs (Fig. 3c), whereas it was not modulated on immature DCs. Finally, as demonstrated in Fig. 3(d), LTC4 significantly stimulated the production of IL-12p40 by LPS-stimulated DCs. Taking into account that p40 is a chain shared by the cytokines IL-12 and IL-23 and the finding that IL-12p70 was strongly inhibited by LTC4, we decided to evaluate the presence of IL-23 in

supernatants of DCs. As shown in Fig. 4(e), LTC4 increased the release of IL-23 in LPS-stimulated DCs, a cytokine associated with the maintenance of Th17

profiles.38,39 The CysLTs exert their effects in several tissues through their action on CysLT1 and CysLT2 receptors.18 Expression Urease of CysLTR1 has been demonstrated in murine DCs.40 Our objective was to evaluate the expression of both receptors in immature and LPS-stimulated DCs by reverse transcription (RT-) PCR. For that, DCs were incubated without or with LPS (1 μg/ml) at 37°, after 30 min we added or not 10–8 m LTC4 and cells were cultured overnight at 37° and finally we analysed the expression of both receptors using RT-PCR. The RT-PCR amplification yielded DNA fragments of the expected size for both CysLTR1 and CysLTR2 (Fig. 4a). By analysis of bands compared with β-actin, we found similar expression for both receptors in immature and LPS-stimulated DCs (Fig. 4b), an interesting fact was that, LTC4 treatment of immature DCs up-regulated the expression of CysLTR1 mRNA. This could suggest that the effects of LTC4 are mediated through the CysLTR1. However, when we analysed DX uptake and cytokine secretion in the presence of montelukast (MK-571), an antagonist of CysLTR1, we found that DX endocytosis only decreased the mean fluorescence intensity in immature DCs by 25–30% (control: 78·2 ± 8·1; LTC4: 165·5 ± 12·4 versus MK-571: 91 ± 15·1; MK-571 + LTC4: 108 ± 21·0, mean ± SEM, n = 3, P < 0·05).

(reviewed in ref 35) It is therefore possible that IL-10, produc

(reviewed in ref. 35) It is therefore possible that IL-10, produced by a small number of skin-resident Treg cells, mediates potent anti-inflammatory effects by serving to limit the amplification of inflammatory networks. With this in mind it is

therefore tempting to speculate that in our model, IL-10 produced by skin-resident Treg cells, acts to suppress the accumulation and survival of neutrophils at the site of antigenic challenge thereby reducing the overall immunogenicity of the antigen. These findings have implications for vaccine efficacy because they indicate that even partial removal of Treg cells will alter vaccine immunogenicity through limiting the influence of the cells on both innate and adaptive immune responses. This work was supported by an MRC non-clinical

GDC-0449 senior fellowship (G117/488), an MRC collaboration grant (G0500617) and project grants from the AICR (05-028) and the Wellcome Trust (067046). The authors declare that there are no conflicts of interest. “
“Membrane microdomains play an important role in the regulation of natural killer (NK) cell activities. These cholesterol-rich membrane domains are enriched at the activating immunological synapse and several activating NK-cell receptors are known to localize to membrane microdomains upon selleck chemicals llc receptor engagement. In contrast, inhibitory receptors do not localize in these specialized membrane domains. In addition, the functional competence of educated NK cells correlates with a confinement of activating receptors in membrane microdomains. However, the molecular basis for this confinement is unknown. Here we investigate the structural requirements for the recruitment of the human activating NK-cell receptors NKG2D and 2B4 to detergent-resistant membrane fractions in the murine BA/F3 cell line an in the human NK-cell line NKL. This stimulation-dependent recruitment occurred

independently of the intracellular domains of the receptors. However, either interfering with the association between NKG2D and DAP10, or mutating the transmembrane region of 2B4 impacted the recruitment of the receptors to detergent-resistant however membrane fractions and modulated the function of 2B4 in NK cells. Our data suggest a potential interaction between the transmembrane region of NK-cell receptors and membrane lipids as a molecular mechanism involved in determining the membrane confinement of activating NK-cell receptors. This article is protected by copyright. All rights reserved “
“Immunoinflammatory-mediated demyelination, the main pathological feature of multiple sclerosis (MS), is regularly accompanied by neurodegenerative processes, mostly in the form of axonal degeneration, which could be initiated by glutamate excitotoxicity. In the current study, the relationship between Th17-mediated inflammatory and excitotoxic events was investigated during an active phase of MS.

Anti-human CD14, CD11b, CD11c, HLA-DR and the respective isotype

Anti-human CD14, CD11b, CD11c, HLA-DR and the respective isotype controls were purchased from

BD (BD biosciences). Anti-human CD86, CD80, CD83 and anti-mouse MHCII were purchased from eBioscience (San Diego, CA, USA). The IL-12p70 ELISA kit was obtained from R&D Systems, and samples were run according to the manufacturer’s instructions. The data in the figures are presented as the mean of quadruplicate wells ± SEM for the mouse BMDCs and triplicate wells ± SEM for MoDCs, respectively. Solubilized antigens as well as the antigenic peptides were prepared as previously described (22). Oocyst excystation (sporozoite preparation) was also performed as previously selleck kinase inhibitor described (23). Briefly, purified oocysts (IOWA isolate) were washed free of 2·5% aqueous potassium dichromate (K2Cr2O7, a storage buffer) with phosphate-buffered saline (PBS, pH 7·4) by centrifugation. Oocysts were resuspended in Dulbecco’s modified Eagle’s medium Tipifarnib mouse base with 0·75% sodium taurocholate and incubated for 15 min at 37°C. The excystation mixture was diluted with Ultraculture™ medium (Lonza Walkersville Inc., Walkersville, MD, USA) and centrifuged

at 18,300 g. The rCp23 (22), rCp40 (22), rCp17 (18) and rCpP2 (19,24) proteins were fused to a Schistosoma japonicum glutathione-S-transferase (GST) tag expressed from plasmid pGex4T-2 in Escherichia coli BL21 cells following the manufacturer’s instructions. The GST fusion tag was cleaved with thrombin (GE Healthcare, Piscataway, NJ, USA), and then, thrombin was removed using pAmino Benzamidine-Agarose (SIGMA # A7155). Endotoxin

was removed using Detoxi-Gel Endotoxin Removing Columns (Thermo Fisher Scientific). rCpP2 was also expressed as a 6 ×  His fusion protein in pQE81 vector (Qiagen, Valencia, CA, USA) using E. coli DH5α Parvulin cells (Invitrogen, Carlsbad, CA, USA) and purified as previously described (19,24). Protein concentrations were determined using the Micro BCA Protein assay (Thermo Fisher Scientific). Endotoxin testing was performed using the limulus amebocyte lysate (LAL), PYROGENT 03 Plus kit, Lonza, according to the manufacturer’s instructions. The lowest limit of endotoxin detection as recommended by the company was set at 0·03 EU. The cells were collected and re-plated in 48-well plates, 200 000 cells/250 μL/well media. Cells were then incubated with either 500 000 sporozoites (approximately 1 : 2 ratio) or different concentrations of antigen for 18 h, after which the culture media were harvested and stored at −80°C for ELISA. Data are expressed as mean ± standard error. ELISA data were transformed and analysed by Student’s t test and one-way anova using Prism software (GraphPad Software, Inc., La Jolla, CA, USA). Luminex data were analysed using MasterPlexTM CT and QT acquire 1.0 and quantitation 2.0 software (Hitachi Solutions, USA). Statistical significance is indicated in the study as *P < 0·05, **P < 0·01, ***P < 0·001. P < 0·05 was considered significant.

The outstanding scientific programme will include plenary session

The outstanding scientific programme will include plenary sessions on fungal infections in all aspects of immunocompromised hosts chaired by an internationally renowned faculty, round table sessions, and meet-the-expert sessions. The poster session will encourage one-to-one discussions between faculty, presenters and delegates. The meeting is designed for infectious disease specialists, haematologists, oncologists, transplant physicians, microbiologists, intensivists, immunologists, dermatologists, paediatricians, learn more and all those with interests in medical mycology.

At the end of the meeting we hope that every participant has learned something new, has been refreshed on something old and has had the opportunity to meet other colleagues within the field of medical mycology. The venue for TIMM-6 is FK506 located in Copenhagen, Denmark. Copenhagen is a vibrant metropolis, the gateway to Scandinavia and amongst the safest and cleanest cities in the world. This beautiful city by the water offers both a wide variety of cultural experiences and stunning architecture within its compact city

centre. Medieval townhouses in a variety of colours and atmospheric streetlamps reflecting in the cobble stones await your delegates in the old city centre. Denmark is the oldest monarchy in the world. Beautiful traces of Copenhagen’s 1,000-year history are to be found everywhere. Through the years, however, Copenhagen has transformed itself into one of the world’s leading design capitals. Award-winning contemporary architecture and stunning design appear all over the city. The Copenhagen Night of Culture 2013 will on 11 October present a sensational programme for all tastes. Museums, to libraries, educational establishments, theatres, musical venues, churches and many other institutions representing art and culture will keep their doors open during the evening from six o’clock to midnight

or beyond. Many of Culture Night’s 500 events are being arranged specially for this evening offering you an experience out of the ordinary. We expect TIMM-6 to be at least as successful as previous TIMM congresses, which brought together more than 1,000 international delegates from all over the world. We look forward to greeting you here in Copenhagen and discuss new developments in medical mycology! Maiken Cavling Arendrup, Cornelia Lass-Flörl, Ditte Marie Saunte and Paul Verweij TIMM-6 Executive Committee “
“We report a case of disseminated fusariosis in an 8-year-old boy with acute myelogenous leukaemia that occurred whilst the patient was severely neutropenic after high-dose chemotherapy. Lung involvement was associated with recurrent typical skin lesions.

Recently, faecal-TB PCR test targeting IS6110 has also been docum

Recently, faecal-TB PCR test targeting IS6110 has also been documented by Balamurugan et al. (2010) in differentiating these two diseases. However, clinical utility of this PCR test is not validated in large number of patients. One major drawback of conventional PCR is that it requires tissue destruction and nucleic acid extraction making impossible click here correlation with histological characteristics (Almadi et al., 2009). An in situ PCR has been developed where IS6110 target was amplified within the intact cells and that combined the ability to localize specific DNA within tissues (Pulimood

et al., 2008). This method could also differentiate intestinal TB from Crohn’s disease in archival mucosal biopsy specimens. However, the sensitivity of in situ PCR needs to be improved and studies should be carried out on large number of patients with Crohn’s disease and intestinal TB before its usefulness is confirmed (Pulimood et al., 2008; Almadi et al., 2009). Cutaneous TB constitutes about 1.5% of all EPTB

cases (Singal & Sonthalia, 2010). However, this disease has re-emerged during the last two decades together with high incidence of PTB and multiple-drug resistant TB (MDR-TB; Abdalla et al., 2009). Differentiation of cutaneous TB from other infectious granulomas of the skin (sarcoidosis, leprosy, fungal SCH727965 or NTM infections) is difficult because of insufficient AFB in the tissue biopsies (Bravo & Gotuzzo, 2007). Of all the clinical types, scrofuloderma is the most commonly encountered variant followed by lupus vulgaris, TB verrucosa cutis and lichen scrofulosorum (Singal & Sonthalia, 2010). Vildagliptin These clinical types

of cutaneous TB have been confirmed by PCR, while smear microscopy and culture test completely failed (Padmavathy et al., 2003). Interestingly, Okazaki et al. (2005) reported first case of M. bovis BCG-derived cutaneous TB (localized at different area from the vaccination site) without immune deficiency by multiplex PCR assay based on region of difference (RD)1, complement sequence of RD1, RD2, RD8, RD14 and SenX3-RegX3 regions originating from M. bovis BCG Tokyo 172. TB cutis orificialis, a rare manifestation of cutaneous TB (caused by auto-inoculation of M. tuberculosis in patients with advanced internal TB), has been confirmed by PCR (Choi et al., 2009). Using culture/histopathology as the gold standard, IS6110-based conventional PCR/nested PCR has been well documented in diagnosing cutaneous TB and that showed superiority over 16S rRNA gene-based PCR (Ogusku et al., 2003; Obieta et al., 2010). A highly sensitive and specific PCR assay targeting 65 kDa protein gene has also been developed for the diagnosis of cutaneous TB, considering culture/response to ATT as the gold standard (Negi et al., 2005a; Abdalla et al., 2009). Ocular TB represents a rare form of EPTB, which accounts for 0.

[7] The klotho knockout mouse is now an established animal model

[7] The klotho knockout mouse is now an established animal model of ageing, allowing further study of well-accepted processes that occur with ageing, such as arteriosclerosis, arterial calcification RAD001 in vitro and osteoporosis, and other less well-studied processes such as angiogenesis.[7, 11, 12] The klotho gene encodes a 1012 amino acid long single-pass transmembrane protein,[7] commonly referred

to as α-klotho, to differentiate it from two subsequently discovered members of the klotho family; β-klotho and γ-klotho. All three are single-pass transmembrane proteins of different lengths, which not only share a substantial degree of homology, but function as obligate co-receptors to endocrine FGF.[13] Within the extensive superfamily of FGF, only the FGF19 subfamily consisting of FGF19, FGF21 and FGF23 are endocrine FGF while the other FGF function as paracrine/autocrine factors.[13, 14] FGF receptors (FGFR) are detected ubiquitously while klotho expression is limited to certain tissues, thereby determining tissue specificity for the endocrine action of their respective FGF.[15] α-klotho is an obligate co-receptor for physiological FGF23

signalling and appears essential for FGF23-mediated phosphate regulation Selleckchem SCH727965 in animal models.[15-17] It is now also evident that klotho proteins play a role in a range of other metabolic processes.[7, 8, 15, 18-20] β-klotho, that augments FGF19 and FGF21 signalling, is found in liver, gall bladder, pancreas, colon and adipose tissue and participates in bile acid metabolic pathways.[19, 20] γ-klotho is coupled to FGF19 and is found in the eye, adipose and kidney and its function remains cryptic.[13] The remainder of this review focuses on α-klotho and will henceforth be referred to as klotho. Klotho exists in two forms – membrane-bound klotho (mKl) and soluble klotho (sKl). mKl is variably expressed in different tissues including parathyroid, brain, heart and testis with low-level expression Tenofovir molecular weight also detected in the aorta.[7, 21] Klotho is most abundantly described in the kidney with earlier reports focused on distal convoluted tubule expression,[7] though more recently

proximal tubule expression of mKl has been reported.[22] sKl, on the other hand, is produced in two ways. The first is a result of ectodomain cleavage of mKl (∼130 kDa) although factors regulating ectodomain shedding remain poorly characterized. A number of proteases have been implicated, most notably a disintegrin and metalloproteinase (ADAM) 10/17, which is also expressed in the distal convoluted tubule. The second is a product of alternative splicing leading to a shorter form of sKl (∼70 kDa). Proteomic analysis of various extracellular fluids suggests that the longer form of sKl, generated by cleavage is the major circulating species in humans.[23-25] The actions of mKl and sKl differ, with mKl predominantly supporting FGF23 in regulating phosphate.

Hierarchical cluster delineation results were validated using non

Hierarchical cluster delineation results were validated using non-hierarchical cluster analysis (kappa inter-classification comparison agreement value κ=0.98). We conclude that this type of analysis can be used to objectively delineate T-cell clusters sharing identical features. We then attempted to determine,

using this approach, whether IL-22-secreting cells are more similar to the Th1 or Th17 subset. As shown in Fig. 2B, the branching point at which IFN-γ-secreting cells are parted from IL-17A- and/or IL-22-secreting cells is more distant from the extremity of the tree, as compared with the branching at which the latter are split into two subsets. As the magnitude of the distance for a given branch point separating two given clusters is directly correlated

with their degree of phenotypical Venetoclax cost differences, Th22 cells appear more closely related to Th17 than to Th1 cells, in PBMCs from the healthy individual taken as an example (Fig. 2B). To confirm this observation, cluster analysis was repeated using PBMCs from a series of healthy (n=12) and psoriasis (n=12) individuals. The results from this analysis confirmed that, in both groups, the distance of the branching point segregating the Th17 and Th22 subsets is significantly shorter than the distance segregating Th1 and any of the latter two subsets (Fig. 2D). Additional parameters (IL-2, TNF-α and CD161) were introduced in order to test their influence on the analysis. As shown in Fig. 2E, AUY-922 in vivo the global clustering pattern was conserved when six parameters were used, except for Th1 cells, which were grouped Sucrase into two distinct clusters

according to their capacity to secrete IL-2 or not. Altogether, six major clusters were defined using six parameters. This result further confirms the restricted number of dominant T-cell subsets sharing identical features, since here sixty-four (26) different clusters could theoretically have been delineated. According to this analysis, IFN-γ+IL-2+ cells would phenotypically be more related to IL-17A- and IL-22-secreting cells, than IFN-γ+IL-2− producers. Of note, the IL-17A and IL-22 parameters were found to cluster together and, importantly, away from IFN-γ. The same pattern was repeatedly observed in 20 out of 24 individuals analyzed (data not shown). Thus, Th17 and Th22 subsets are distinguishable and defined as separate entities, even when a more complex analysis is performed. As shown above, IL-17A- and IL-22-secreting cells are relatively scarce in periphery, even in psoriasis patients (Fig. 1 and Supporting Information Fig. S1). To determine whether these cells are more abundant in inflamed tissue lesions, infiltrating T cells were expanded in vitro from both healthy skin and psoriasis lesions of the same patients (n=3) and their cytokine production profiles analyzed by multiparametric flow cytometry (Supporting Information Fig. S3A).

After centrifugation

of the purified bacteria, 1 mL of th

After centrifugation

of the purified bacteria, 1 mL of the pellet [0.8 × 106 DNA copies of FAM cycle threshold (Ct) at 21.41] was used to infect fresh RG7204 nmr XTC-2, Vero or L929 cells (control) for 1 h at room temperature. Then, 4 mL of either fresh L-15M medium (5% FBS) or MEM (4% FBS) that was either supplemented with (2%) TPB or unsupplemented (growth control) was added to the culture flasks containing each cell line. All of the inoculated flasks were incubated at 28 °C. The medium was replaced every week with fresh medium (L-15M or MEM with or without TPB) for 3 weeks to ensure adequate nutrition. Finally, to verify that R. felis was successfully maintained in Vero and L929 cells, subpassages were performed every 3 weeks. Each experiment was performed at least twice. The viability and growth rate of R. felis in XTC-2, Vero and L929 cells were detected using quantitative PCR (qPCR), Gimenez staining and indirect fluorescent-antibody (IFA) staining of cytospin preparations. For qPCR, a specific probe (6FAM-AGGTGATGGAGAGGTTACCGGTGGAG-TAMRA) and a primer pair (Fwd: 5′-CCGTTGCCGGTAGCTTGTAT-3′; Rev: 5′-GCATTTGCAGCCCCCTCTAT-3′) were designed to target the cell surface antigen-like protein Sca7 gene of R. felis. Relative quantification of the qPCR results was performed as described by Mba et al. (2011). For IFA staining, a mouse monoclonal

Selleckchem ABT-263 antibody against R. felis was used to confirm the intracellular growth of R. felis, which was monitored by Gimenez staining. Comparative analysis of R. felis replication was investigated in amphibian Molecular motor and mammalian cells inoculated with R. felis species in two culture media, L-15M and MEM, which were unsupplemented or supplemented with TPB. Using L-15M:TPB medium, qPCR, Gimenez and IFA results showed that R. felis replicated better in XTC-2 cells than in either mammalian cell line on day 7 (50 × 106 DNA copies), but both XTC-2 and L929 cells enabled better R. felis growth

(40–50 × 106 DNA copies) than did Vero cells (9 × 106 DNA copies) (Table 1, Supporting Information, Fig. S1) on day 14. A cytopathic effect of infected cells was observed for Vero and L929 cells during the third and second weeks, respectively, using inverted phase contrast microscopy (Fig. S2). When using MEM:TPB medium, qPCR showed that R. felis growth was similar in Vero and L929 cells after 7 days whereas at day 14, R. felis growth was greater in L929 cells (50 × 106 DNA copies) than in Vero cells (10 × 106 DNA copies) (Table 1). Overall, R. felis growth was similar in Vero and L929 cells in both MEM:TPB medium and L-15M:TPB medium at day 7, but R. felis grew better in L929 cells than in Vero cells at day 14 in both media and grew to similar levels in L929 cells and XTC-2 cells in L-15M:TPB medium (Table 1). Using media with and without TPB, we found a positive effect of TPB on R.

Although level of pVL is closely associated with the rate of HIV

Although level of pVL is closely associated with the rate of HIV disease progression, it does not measure disease progression directly. We therefore calculated the rate

of decline in CD4+ T cell counts (see the Materials and Methods), and investigated their association with HLA allele expression as well, but failed to detect any alterations in the rate of decline as the HIV epidemic matured (data not shown). This may be due to the low statistical power of the present study, therefore larger scale studies are warranted in order to determine to what extent, and for which HLA alleles, such accumulations of CTL escape have been occurring, and how they have been affecting disease Alectinib chemical structure progression. In the present study, we have Y-27632 solubility dmso demonstrated that: (1) there are no individual HLA class I alleles which are strongly associated with the level of pVL in the Japanese population at the current time; (2) the Japanese population has a narrow HLA distribution and lacks in the most protective HLA-B27/B57; (3) the proposed advantage of rare class I supertypes and the disadvantage of homozygotes

for Bw6 motif cannot be applied to all ethnic groups across the globe; and (4) HLA-B51 has been losing its dominant effects at the population level over time, whereas this is not the case for the other alleles. Despite substantial numbers of HIV-1 viremia controllers having been recognized in Japan, this population lacks the well-known protective alleles HLA-B27/B57. We therefore expected to discover novel associations between HIV disease progression and HLA class I alleles which are unique to Asian populations. However, in the cross-sectional analysis, we did not identify any significant associations between the level of pVL and expression of individual class I alleles, indicating

that, regardless of the geographical part of the world, the protective effects of HLA alleles are greatly biased to a few of the prominent alleles like HLA-B27/B57. The discordant results for HLA supertypes and homozygosity of the Bw6 motif between Japan and the USA are likely Montelukast Sodium also attributable to the lack of HLA-B27/B57 in the Japanese population. These two exceptional alleles are known to have targeting epitopes within Gag protein (10, 30–35). Likewise it has been suggested that expression of HLA alleles other than B27/57, but having targeting epitopes within Gag protein, are associated with lower pVL (8, 36–40). Therefore it is warranted to confirm that Gag specific CTL responses are associated with lower pVL in Japanese people who lack HLA-B27/57. In the cross-sectional analysis, we did not identify significant associations between pVL and HLA-A11, 26, B51 or Cw14 expression, all of which have been shown to be protective in Caucasians (7), However, subsequent analysis revealed that HLA-B51, at least, was protective in the past, indicating that there has been loss of targeting epitopes in the viral strains circulating in this population.

As illustrated in Fig 4E, the addition of CXCR3+ CD25hi cells in

As illustrated in Fig. 4E, the addition of CXCR3+ CD25hi cells into the cultures in increasing ratios suppressed proliferative responses to baseline. Taken together, these observations indicate that subset(s) of CXCR3-expressing T cells have potent immunoregulatory properties. We next evaluated the functional implications

of CXCR3 Quizartinib expression on Tregs for IP-10-dependent chemotaxis. Leukocyte migration was measured using a microfluidic technique that allows for precise and robust measurements of leukocyte migration at single-cell resolution 46. Purified CD4+CD25+ CD127dim/− Tregs were FACS-sorted into CXCR3pos or CXCR3neg subsets and were introduced into the main channel of the microfluidic device (Fig. 5A). Subsequently, images of live-time cell migration toward the chemokine IP-10 selleck compound were captured using time-lapse imaging, as described in Materials and methods. In the absence of a chemoattractant stimulus, we found minimal migration of T cells into the 6×6 μm side channels, and cells that entered the channels appeared to move at random.

However, as illustrated in Fig. 5B and C, we found that CXCR3+ Tregs had a marked chemotactic response toward IP-10, and their directional persistence was significantly greater (p<0.01) than that observed for CXCR3neg Tregs (Fig. 5D). CXCR3neg subsets were found to move in a random manner, some cells entered the channel and returned to baseline, and some migrated toward IP-10. In general, the directional persistence of CXCR3neg subsets was limited (Fig. 5D). We also observed that the velocity

of CXCR3pos cells during persistent directional migration was consistently slower than the velocity of random migrating CXCR3neg Tregs (but this difference did not reach statistical significance, data not shown). Collectively, these studies demonstrate that CXCR3 is functional to elicit chemotaxis in CXCR3-expressing Tregs. We next wished to evaluate the co-expression of CXCR3 with well-established lymphoid and peripheral homing receptors on FOXP3+ Tregs. We stained PBMC for CD4, CD25, FOXP3 and either CXCR3, CD62L, CCR4, CCR5 and CCR7, established to be expressed on Tregs 22–26. We also evaluated the co-expression of CXCR3 4��8C with Treg-associated homing receptors. Illustrated in Fig. 6A and B, we found comparable levels of CXCR3 and CD62L expression on both CD25hiFOXP3+ Tregs and CD25loFOXP3− Teff subsets. However, among chemokine receptors, we found lower levels of expression of CCR7 and higher levels of CCR4 and CCR5 on FOXP3+ Tregs versus Teff subsets. Also, we observed that CXCR3 is co-expressed with CD62L on ∼30% of FOXP3+ Tregs, while only ∼12% Tregs co-express CCR7 and CXCR3; and ∼20% CXCR3pos Tregs co-express CCR4 or CCR5 (Fig. 6B).