Ching and colleagues have developed a rapid immunochromatographic

Ching and colleagues have developed a rapid immunochromatographic flow test to detect the anti-O. tsutsugamushi IgG and IgM in patients’ sera for diagnosis of scrub typhus, by employing a Karp r56 protein that contained deletions of 79 and 77 amino acid residues at the N and C terminals, respectively, as the diagnostic antigen (19, 20). Antibodies prepared from serum of patients with scrub typhus tend to recognize this protein in general. Mice immunized with the 56-kDa protein generated neutralizing antibodies and showed increased resistance to homologous O. tsutsugamushi infection (21). These data suggest that it is a favorable diagnostic antigen and

vaccine candidate. In this report, we describe the Daporinad cost molecular cloning, expression and purification of the 56-kDa protein from O. tsutsugamushi strain Karp and investigate the immunogenicity of the recombinant protein. Primers were designed based on the Selumetinib research buy published 56-kDa gene nucleotide sequence (GenBank accession no. M33004.1). The upstream and downstream primers were designed to contain NcoI and XhoI restriction sites, respectively: Ot56-F

(positions 298–316), 5′-AGACCATGGCTCAGGTTGAAGAAGGTA-3′; and Ot56-R (positions 1386–1404), 5′-GTCTCGAGCTAAGTATAAGCTAACCCT-3′. Genomic DNA isolated from O. tsutsugamushi strain Karp was used as a template. PCR was performed in a final volume of 50 μL containing approximately 50 ng DNA, 200 μM each deoxyribonucleotide triphosphate, 10 pmol each primer, 5 μL of 10 × PCR buffer (Mg2+ Plus; TaKaRa Biotechnology, Dalian, China) Amisulpride and 0.5 U of Ex-Taq DNA polymerase (Takara Biotechnology). Thermal cycling conditions were as follows: 2 min at 95°C, 2 min at 95°C, followed by 30 cycles of 30 s at 94°C, 30 s at 57°C and 1 min at 72°C. A final step of 10 min at 72°C was added to the last cycle. PCR products were analyzed by 1% agarose gel electrophoresis. pET30a(+) and purified PCR products were digested with restriction enzymes NcoI and XhoI (TaKaRa Biotechnology), then ligated overnight at

16°C. The ligation mixture was initially introduced into E. coli DH5α. The recombinant plasmids were identified by PCR, enzyme digestion and were confirmed by sequencing. The plasmid construct was then transformed into E. coli Rossetta (Novagen, Madison, WI, USA) for expression. Escherichia coli Rossetta containing the appropriate plasmid was cultured at 37°C in LB broth containing kanamycin and chloramphenicol. Cultures were induced at an OD600 of 0.6–0.7 with IPTG to a final concentration of 1 mM, and grown for a further 5 hrs. Cells were then pelleted and resuspended in 50 mM phosphate buffer (pH 7.4). After cell lysis by sonication, cellular debris were eliminated by centrifugation at 8000 g for 15 min at 4°C. The water-soluble fraction of the lysate was collected for purification, as described below. To purify the recombinant protein, the cell lysate, containing protein with six His tags, was filtered through a 0.

The prevalence of CKD in Australian adults is approximately 16% w

The prevalence of CKD in Australian adults is approximately 16% with 2.4% having proteinuria and 7.8% CKD stages 3–5.25 Considering general untargeted screening of the population is not supported in Australia for its ineffective manner,7 the study demonstrated that early detection and optimal management of high blood pressure, diabetes and proteinuria in a primary care-based setting incorporating annual screening in 50–69 year olds, along with intensification of management in those already Z-VAD-FMK cell line known to have these conditions, would be cost-effective and in some cases

highly cost-effective. Particular benefits of such a program, incorporated into an existing primary care system, lay in reducing cardiovascular and ESRD deaths, as well as reducing the number of people needing dialysis or transplantation.26 Another approach of opportunistic primary care-based targeted screening of high-risk

individuals is to conduct similar GSK 3 inhibitor targeted screening programs in the community. A community-based detection program has been developed by Kidney Health Australia and piloted in the Australian workplace environment. Entitled KEY (Kidney Evaluation for You), the objectives were to test an effective and affordable means of finding early asymptomatic CKD in high-risk individuals within the community and referring them to a primary health-care provider for appropriate long-term care. The GNAT2 pilot studies have shown promising detection rates, however, further development of the KEY program and expansion into other community sites such as pharmacies and workplaces will depend on cost–benefit analysis. The most sustainable and effective approach appears to be opportunistic general practice screening, with the emphasis on early detection. The well-identified screening process of blood pressure, estimated GFR (eGFR) and urinary protein fits well with the developing approach to chronic disease, particularly given the ease of identification of the high-risk

groups, the simple tests needed to establish the presence and staging of CKD and the overlap of the action plans for CKD with those for best care of people with diabetes and cardiovascular risk reduction. However, for early detection and management of CKD to be successful in reducing the growing burden of CKD, substantial effort at education within primary care is required and subsequent treatment regimens will need to be broad-based for chronic disease management as a whole, and made cost-effective for the practitioner. Early detection of CKD is important for prevention and control of the disease. Studying the cost-effectiveness of the CKD prevention program may facilitate better management of the disease.

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 Bioactive Compound Library 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

see more 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 Morin Hydrate 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.

Human pDCs secrete high levels of IFN-α in response to TLR7/8-L a

Human pDCs secrete high levels of IFN-α in response to TLR7/8-L and CpG class A and C while other cells show no or low detectable amounts of IFN-α.2,3,25,32 Because pDCs are rare cells in the immune system, direct isolation to study these cells in detail requires large volumes of blood. To compare IFN-α secretion in rhesus and human pDCs we therefore used the staining panel presented above for identification of these cells out of total PBMCs. As the objective of the present study was to compare pDC-mediated enhancement of B-cell responses, we only

compared the IFN-α production with the ligands that also induce B-cell proliferation, i.e. CpG C and TLR7/8-L here. Hence, PBMCs were stimulated Fulvestrant molecular weight for 12 hr with CpG C or TLR7/8-L, intracellularly stained for IFN-α production in CD123+ pDCs and analysed by flow cytometry. In both rhesus and human

cultures, IFN-α-secreting pDCs were detected in response to CpG C and TLR7/8-L. Markedly higher frequencies of producing selleck inhibitor cells were observed in response to TLR7/8-L (Fig. 3a). No IFN-α expression was detected by flow cytometric intracellular staining in any other cell population than CD123+ pDCs (data not shown). We previously reported that a large proportion of human pDCs display a rapid IFN-α secretion on a per cell basis after TLR7/8-L stimulation and that other stimuli such as virus exposure exhibit delayed kinetics where the IFN-α levels accumulate over time.34 Although virus exposure may be different from stimulation with single TLR ligands, we observed a similar phenomenon where the supernatants from parallel rhesus and human cultures harvested at 24 hr and analysed Resminostat by ELISA showed that the levels of IFN-α induced by CpG C exceeded

the levels found by TLR7/8-L (Fig. 3b). This effect was more pronounced in the human cultures (P = 0·001) than in the rhesus cultures (P = 0·556). When comparing the absolute IFN-α levels between human and rhesus cultures, CpG C was shown to induce higher levels in the human cultures whereas TLR7/8-L induced higher levels in the rhesus cultures (Fig. 3c). Since the detection reagents used in both methods are reported to be cross-reactive between rhesus and human IFN-α, we concluded from these data that although human and rhesus pDCs produce IFN-α in response to both TLR7/8-L and CpG C, the levels and kinetics appear to differ. Emerging data indicate that pDCs via production of IFN-α play an important role in shaping the humoral immune response induced by virus infections or vaccination. Human B-cell proliferation and differentiation into antibody-producing plasmablasts in response to TLR7/8 ligation were shown to be significantly augmented by IFN-α produced by pDCs.

7a) Induction of IL12p40 expression on rhesus pDC, as observed w

7a). Induction of IL12p40 expression on rhesus pDC, as observed with mAb C8·6, was confirmed by using another anti-IL-12p40/70 mAb (clone

C11·5), which gave similar percentages of positive cells for pDC as well as mDC upon TLR-7/8 stimulation (Fig. 7b). Finally, analysis of IL-12p40 mRNA in TLR-7/8 (CL097)-stimulated purified pDC, mDC and monocyte populations showed similar high expression levels in pDC relative to mDC and monocytes and no induction in pDC upon TLR-4 stimulation (Fig. 8a), thus confirming the FACS expression data. Both mDC and monocytes up-regulated TNF-α mRNA expression upon TLR-4 (LPS) as well as TLR-7/8 (CL097) stimulation, underscoring the functional capacity of these purified cell populations (Fig. 8b). In agreement with the FACS analysis, TNF-α mRNA expression in pDC was up-regulated only upon TLR-7/8 and not TLR-4 stimulation. While the mDC and pDC preparations were only 60–75% pure the other cells present were PF-02341066 solubility dmso www.selleckchem.com/products/dabrafenib-gsk2118436.html either granulocytes or monocytes

and this could not have affected the IL-12p40 expression data, as monocytes were observed to have only very low IL-12p40 expression and the monocyte fraction itself was >90% pure with <5% mDC and <1% pDC contamination. In this work, we adapted a whole blood stimulation assay to study functional characteristics of peripheral blood DCs and monocytes in macaques and performed a direct comparison with human blood samples. Most responses of the different subsets were very similar between macaques and humans and in agreement with previous studies, in which purified cell populations instead of whole blood stimulation had been used [2, 17, 25-28, 32]. However, we observed that, in contrast to humans, rhesus pDC expressed

IL-12p40 upon stimulation with TLR-7/8 or TLR-9. Preliminary data suggest a similar IL-12p40 expression pattern in cynomolgus macaques (V.S., to be published elsewhere). We also observed that relative to humans, mDC why and monocytes in rhesus macaques responded less well to TLR-7/8 stimulation when expressed as percentage of IL-12p40- and TNF-α-positive cells. Of note is that a similar relatively lower level of IL-12 induction has been reported previously for macaque monocyte-derived DC [23]. The capacity of rhesus pDC to produce IFN-α as well as IL-12p40 may potentially modify their response to viral infections, where pDC are known to play an important role [36]. Previous studies either did not include IL-12 in their analysis [23] or measured IL-12 cytokine production by enzyme-linked immunosorbent assay (ELISA) on either stimulated total PBMC or lineage-negative cell cultures [25-27]. Others used FACS analysis, but studied IL-12 expression only in LPS-stimulated PBMC [17], which would have given no expression in pDC. Hence, our observation was made possible by the use of FACS analysis to detect TLR-induced cytokine expression in all subsets simultaneously.

Indeed, as the subtle nuances of the intimate developmental relat

Indeed, as the subtle nuances of the intimate developmental relationships between T cell subsets continue to emerge [23,24] it becomes apparent that Tregs are not equally suppressive of all subsets or the functions thereof. In fact, in certain circumstances Tregs can promote and potentially stabilize the Th17 developmental programme [6], thus fully warranting their description as ‘regulatory’ selleck chemicals rather than simply ‘suppressor’ cells. It appears

that FoxP3 can protect against pathology at various levels. Technological advances, in particular the generation of FoxP3 and RORγt reporter mice [15,25], have provided greater finesse, allowing the unequivocal identification of iTregs[26–28] and dissection of the lineage relationships between iTregs and Th17 cells [5]. These experiments therefore identified the possibility that ‘suppression’ could not only be mediated via the action of established Tregs on responder cells, but could also operate at the level of lineage commitment. Mice with conditional cell-specific deficiencies in targeted elements of the suppressive machinery used by Tregs are now allowing the relative importance of these elements to be addressed with increased precision [29–31]. For example, FoxP3 can interact directly with elements involved in both Th17 (RORγt) and Th2 interferon regulatory factor-4 (Irf-4) lineage commitment

[25,32]. Thus FoxP3 can act to suppress inflammation directly, by physically preventing the activation of proinflammatory programmes in the cell in which it is expressed. The TCR repertoire of Tregs is selleck products thought to be enriched for self-reactive TCRs [33]. Therefore,

Tregs may represent a significant pool of autoreactive cells if they were able to gain proinflammatory effector function. Bearing this in mind, it is unclear whether the pathologies seen 5-FU datasheet in the scurfy mutant or FoxP3 knock-out mouse reflect a gain of effector function by ‘Tregs’ expressing non-functional FoxP3 or from the activation of self-reactive naive T cells from the FoxP3– peripheral repertoire. Selective depletion of FoxP3-expressing cells can be achieved by administering diphtheria toxin to mice engineered to express the human diphtheria toxin receptor in FoxP3+ cells [34]. Treg depletion via this system induced the rapid onset of fatal autoimmune disease, indicating that autoaggressive T cells arising from the FoxP3– pool are sufficient to recapitulate the scurfy phenotype. However, other studies have indicated that there is also pathogenic potential within the Treg compartment. FoxP3 function is not binary in nature, and Tregs expressing an attenuated level of FoxP3 were found to display a reduced expression of Treg‘signature’ genes and an increased propensity to differentiate into Th2 effectors [35].

However, here we concentrate on evidence for differential sensiti

However, here we concentrate on evidence for differential sensitivity as measured by T cell effector functions. Thornton and Shevach described

a co-culture system to measure Treg-mediated suppression that not only provided important mechanistic data on the requirements for suppression, but also laid down a template for demonstrating the functional activity of Tregs. The classical suppression assay involves the co-culture of CD25+ Tregs and CD25– responder T cells over a range of suppressor : responder ratios and measurement of the extent to which Tregs restrain the proliferation of CD25– T cells [40]. There is almost no area of Treg learn more biology which has not been assessed by some modification of this basic technique. This assay has been used to compare the regulatory function of different subsets of Tregs[64], of in vitro-activated versus freshly explanted Tregs[65–68], of Tregs from sites of inflammation [69], of nTregs and iTregs[26] and of Tregs in infected versus healthy mice [70] and humans [71]. The findings of

many of these studies informed further in-vivo experiments and they have greatly enhanced our knowledge of Treg function. However, the specificity and activation status of regulatory and effector T cell populations as well as the cytokines present in the microenvironment and the activation status of antigen-presenting cells (APCs) will influence the capacity of Tregs to suppress in vivo. These conditions are often not well modelled in vitro and this caveat represents the greatest limitation of this type of assay. Particularly in mice, Dabrafenib most often the responder population used for in vitro suppression assays are CD4+CD25– T cells from naive mice, and such cells are highly susceptible to Treg-mediated suppression.

Indeed, it has been suggested that the window of susceptibility to Treg-induced suppression in vitro is regulated tightly and restricted to the first 12 h of stimulation [72]. Limiting the ADAM7 proliferation or cytokine production of highly activated polarized T cells is a much more demanding task, and this may be why a clear comparison of the capacity of Tregs to limit the activity of polarized Th1, Th2 and Th17 cells is missing from the literature. It has been shown, however, that while Tregs can suppress the priming of Th2 responses, they are unable to suppress the proliferation or cytokine production of established Th2 effectors unless they themselves are pre-activated in vitro[73]. The importance of the comparative activation status of effectors and Tregs has been well illustrated. Tregs at sites of inflammation, for example, are typically more highly activated than peripheral Tregs[74,75], and this draws into question the extrapolation of functional assays carried out using mismatched responder : suppressor co-cultures and argues in favour of sampling both Tregs and effector T cells from the tissue of interest wherever possible [44,69,76].

One of the advanced lipid-based delivery systems is the solid–lip

One of the advanced lipid-based delivery systems is the solid–lipid nanoparticles (SLNs), which can be one of the alternative delivery system to electroporation. SLNs are basically composed of high-melting-point lipids that act as a solid core, covered by surfactants. The use of materials that are generally recognized as safe (i.e. triglycerides, partial glycerides, fatty acids, steroids) [35] leads to an advantageous toxicity profile [36].The SLN production by hot high-pressure homogenization is easy, and no organic solvents are required [37]. Scaling-up is standardized up to 50-kg batches [38], and steam sterilization is possible [39]. The excellent activity and superiority of DOTAP–cetyl palmitate–SLN were reproducible.

The positively charged SLN would bind to polyanionic DNA via electrostatic

force leading Ivacaftor price to SLN–DNA complex that will protect DNA from interaction with small molecules in the environment and will be taken into cell by an endocytosis process [40]. An additional advantage of delivering vaccine candidates by nanoparticles is the potential to enhance their stability during transport, and this is critical in areas that lack reliable cold storage chain (2–8°C) [41]. Our previous results revealed that stable formulation of cSLN was able to protect pDNA in DNase I challenge assay and deliver it to the right immune cells for the proper immune response induction [22]. In this study, we generated a DNA vaccine encoding A2–CPA–CPB−CTE as a trifusion gene and compared the impact of DNA vaccine delivery to immune cells (e.g. physical/electroporation vs. chemical/cSLN formulation) on the development of protective immune response against an infectious Tipifarnib ic50 L. infantum challenge. The pcDNA–A2–CPA–CPB−CTE was formulated into cationic

lipid particles with nanometre Parvulin range (~240–250 nm). In our experimental system, the administration of pcDNA–A2–CPA–CPB−CTE in BALB/c mice elicited the induction of specific Th1 and Th2 clones, indicating a mixed immune response and the production of IFN-γ and IL-10, although IFN-γ was much higher than IL-10, especially in G2 using the cSLN formulation. However, a higher amount of IFN-γ was obtained in G1 immunized via electroporation in response to both rA2–rCPA–rCPB and F/T L. infantum antigens at 4 and 8 weeks after challenge. Although IFN-γ secretion at 8 weeks after challenge in G1 was higher than in G2, there were no significant differences in IFN-γ: IL-10 ratio between these two groups. Also, at 8 weeks after challenge, the IFN-γ: IL-10 ratio in splenocytes from mice immunized with pcDNA–A2–CPA–CPB−CTE (G1 and G2) stimulated with rA2–rCPA–rCPB was significantly higher than G3 (~28·25- and 26·5-fold; P < 0·01) and G4 (~8·69- and 8·154-fold; P < 0·01). The same result was obtained with splenocytes stimulated by F/T L. infantum antigen. So, we can conclude that these two delivery strategies elicit the same immune responses with efficient protection.

While autoimmune diseases have been linked with genetic polymorph

While autoimmune diseases have been linked with genetic polymorphisms of co-stimulatory markers [21, 22], the functional Selleckchem Fluorouracil implications have not yet been fully deciphered. Genetic polymorphism,

of course, may compromise not only the function of these molecules but their detection by antibodies. The lack of cell surface CD28 prompted the investigation of the possible expression of alternative co-stimulatory molecules, PD-1, ICOS and 4-1BB, by CD8+CD28− Treg. The expression of all these molecules was higher on RA SF CD8+CD28− cells compared with paired PB Treg, perhaps reflecting the higher activation status of the SF cells. The SF cytokine milieu also contains high local concentrations of IL-15 and IL-12 which down-regulate CD28 but enhance 4-1BB, ICOS and PD-1 expression by CD8+ T cells and increase CD8+ cell survival [23]. CD4+CD25+ Treg display attenuated regulatory function following 4-1BB expression [24]. As 4-1BB expression was reduced in RA(TNFi), this raises the

question as to whether or not it might be a component of the improved suppressor function by CD8+CD28− Treg following therapy in RA(TNFi) patients. The ability to suppress T cell responses may therefore be a balance between the pro-proliferative drive of 4-1BB and the inhibitory effect of other C59 wnt ic50 mediators, such as PD-1. Overall, a relatively low expression of PD-1 and ICOS was shown by all CD8+CD28− Treg samples. Nevertheless, PD-1 has been linked positively to CD8+CD28− Treg with suppressor function in lupus-prone mice [25]. Therefore, it was notable that PD-1 expression by RA(TNFi) was increased compared with RA(MTX), although still below healthy control levels. For further insight into the defective CD8+CD28− Treg in RA, cells were used in cross-over co-culture experiments between the RA(MTX) and HC subjects. RA(MTX) CD8+CD28− Treg remained unable to suppress allogeneic healthy or RA responder

cells, whereas HC CD8+CD28− cells suppressed allogeneic HC responder cells but not RA(MTX) responder T cells. This finding complements the fact that responder T cells had reduced sensitivity to CD4+CD25hi Tregs in active SLE [26] and type 1 diabetes patients [27], suggesting that in autoimmune diseases Treg activity is hampered by both defective Non-specific serine/threonine protein kinase Treg function and the relative insensitivity of the responder cells. The effect of TNF inhibitor on the ex-vivo phenotype and function of CD8+CD28− cells, such as the increase in IL-10R expression on RA(MTX) T cells, suggests strongly that these cells are only temporarily incapacitated by TNF-α and when this is removed from the environment the activity appears to return to normal. However, RA(TNFi) expression of IL-10R remained lower than normal HC expression and suggests that other mediators are involved. Continuing these studies, the role of IL-10 and TGF-β is under further investigation. Longitudinal studies will be performed to address the effect of therapy on CD8+CD28− Treg.

All animal experiments were performed according to institutional

All animal experiments were performed according to institutional guidelines approved by the Niedersächsisches Landesamt

für Verbraucherschutz und Lebensmittelsicherheit. The mAb used for ex vivo iIEL stimulation directed against γδ TCR (clone GL3), CD3 (clone 145-2C11), αβ TCR (clone H57-597) (all Armenian hamster) were purified from hybridoma supernatants and γδ TCR (clone GL4) was a gift from Dr. Leo Lefrançois. For Ca2+-flux studies anti-γδTCR (clone GL3), CD3 (clone 145-2C11) and goat anti-Armenian hamster (anti-Hamster, Jackson ImmunoReasearch) were applied. For the analysis of T-cell populations by FACS the following mAb were used: γδTCR-FITC (clone GL3), γδTCR-biotin (clone GL3) and CD3-biotin

(clone 145-2C11), CD8α-Cy5 or CD8α-biotin (clone Rm CD8), CD8β-Pacific Orange (clone Rm CD8-2), CD4-Pacific Blue (clone GK1.5), CD62L-biotin JQ1 research buy (clone MEL-14) and Fc receptor (clone 2.4G2) were purified from hybridoma supernatants; anti- CD69-biotin (clone H1.2F3) and Streptavidin-PerCP were obtained from BD Bioscience, CD44-biotin (clone IM7) from Caltag and αβ TCR-APC-AlexaFluor 750 (clone H57-597) MK-8669 from eBiosciences. For measurement of intracellular cytokines, we used polyclonal goat anti-mouse CCL4 (R&D Systems), polyclonal F(ab′)2 Donkey anti-goat IgG-PE (Jackson ImmunoReasearch), ChromPure goat IgG (Jackson ImmunoReasearch) or anti-IL-17A-PE (clone ebio17B7, eBiosciences) and anti-IFN-γ-PE (clone XMG1.2, Caltag). iIEL were isolated according to a modification of a previously published method 39. Briefly, the small intestines were flushed with

cold PBS 3% FBS, connective tissue and Peyer’s patches were removed and the intestines opened longitudinally. Next, the small intestines were incubated two times for 15 min in a HBSS 10% FBS 2 mM EDTA at 37°C, shaken vigorously Janus kinase (JAK) for 10 s and cell suspensions were collected and pooled. The cell suspension was filtered through a nylon mesh and centrifuged at 678×g, 20 min at room temperature, in a 40%/70% Percoll (Amersham) gradient. The iIEL were recovered from the interphase and were washed with PBS 10% FBS. Systemic T cells were isolated from systemic lymphocytes of spleens and systemic lymph nodes from γδ reporter mice (F1 C57BL/6-Tcra−/−×TcrdH2BeGFP), mashed in nylon filters, both mixed and subjected to erythrocytes lysis. Next, the cell suspension was washed with PBS 3% FBS, filtered through a nylon mesh and resuspended in RPMI 1640 10% FBS for further analysis. γδ reporter mice were treated with a regime of three consecutive intraperitoneal injections of purified anti-γδ TCR mAb at day −6, day −4 and day −2 before analysis (clone GL3, 200 μg/mouse). Control groups received mock injections with PBS. iIEL and systemic T cells from γδ reporter mice were prepared for Ca2+-flux cytometry as described with minor modifications 58.