Our results indicate that this signalling shift in T cells is tri

Our results indicate that this signalling shift in T cells is triggered due to ligation of low-affinity FcRs by ICs in the presence of TCC. Phosphorylation of ITAM in FcRγ chain is responsible for Syk activation, which then subsequently participate in downstream activation of mitogen-activated protein kinases (MAPKs), PI3K and PLCγ activation in lymphocytes. In order to establish a role for Syk in IC-mediated T cell

activation via low-affinity FcRs, we probed for phosphorylated Syk in the activation loop at Tyr525/526 in cells treated with ICs and TCC. The immunoprecipitates prepared using monoclonal anti-FcγRIIIA/B antibody from cells treated with TCC and ICs, when probed with anti-pSyk, showed phosphorylation of a protein band that migrated at 72 kD. This suggested Syk activation Raf inhibitor in T cells, in response to ICs and TCC (Fig. 2c). These

findings are also supported by our previous observation of Syk phosphorylation in Jurkat cells treated with TCC and in vitro formed ovalbumin–anti-ovalbumin ICs and ICs purified from plasma of SLE patients [26]. These results are also supported by the previous observation that C59 wnt manufacturer Syk is activated in SLE T cells [28]. Syk activation is mediated via FcRγ chain [17]. We observed that in CD4+ T cells treated with ICs or ICs and TCC, the FcRγ chain was recruited to the site of membrane receptors (Fig. 3a). The co-localization analysis of all the Z-series sections (Fig. 3biii) confirmed this finding. The presence of TCC during the IC treatment enhanced the recruitment of the FcRγ chain with membrane FcγRIIIA (Fig. 3biii). Although the observed scatter-pattern for the co-localization of FcRγ chain was different from the pSyk and FcγRIIIA/B staining, we presume that this was due to wider distribution of the staining intensity of the FcγRIIIA and FcγRIIIB receptors, both of which were recognized by the monoclonal antibody that was used for the staining (Fig. 3a). The scattergram obtained in both co-localization out experiments demonstrated data where a line of best fit could be

drawn confirming the association among these proteins. An antibody that recognizes both receptors was used in this study due to the unavailability of an antibody that recognizes only FcγRIIIA. TCC alone was insufficient to trigger these events. The cells stained using anti-FcγIIIA/B antibody demonstrated localized peripheral membrane staining (Fig. 1b). A similar staining pattern was also observed with an affinity-purified anti-FcγRIIIB antibody. Both FcγRIIIA and FcγRIIIB co-localized with labelled AHG on cell membrane (Fig. 1b). Co-staining of expanded naive CD4+ T cells using anti-FcγRIIIA/B and anti-FcγRIIIB demonstrated that those CD4+ T cells that expressed FcγRIIIA always expressed FcγRIIIB.

In conclusion, this study demonstrates that AFP impair the DC abi

In conclusion, this study demonstrates that AFP impair the DC ability of activation of NK cells. These findings might provide new insight into understanding the mechanisms underlying the suppression of innate immune responses

in chronic liver disease patients with high serum AFP levels. This work was supported by a Grant-in-Aid from the Ministry of Education, Culture, Sports, Science and Technology of Japan and a Grant-in-Aid for Research on Hepatitis and BSE from the Ministry of Health, Labour and Welfare of Japan. The authors have no conflicts of interest. “
“Antineutrophil cytoplasm autoantibodies (ANCA) directed against bactericidal/permeability-increasing 3-Methyladenine molecular weight protein (BPI) are common in patients with cystic fibrosis (CF), and serum levels are correlated with lung colonization by Pseudomonas aeruginosa and the severity of lung damage. The production of BPI-ANCA may be due to the costimulation of BPI when mounting an immune response against P. aeruginosa. The effect of surgery aiming to eradicate bacteria and infected tissue on BPI-ANCA levels is sparsely described. A cohort of patients with CF were included: 53 patients having extensive

image-guided sinus surgery (EIGSS) with topical postoperative antibiotic treatment, 131 non-operated controls and 36 who had double lung transplantation (LTX). In all 219 patients, serum samples before and after surgery or at similar intervals were analysed for IgG and IgA BPI-ANCA. The EIGSS group showed a highly significant decrease Talazoparib chemical structure in both IgA and IgG BPI-ANCA levels compared with their own preoperative values and control

group values (P < 0.001–0.02). The LTX patients also showed a highly significant decrease in both IgA and IgG BPI-ANCA levels (P < 0.001). EIGSS and LTX decrease IgA and IgG BPI-ANCA levels in patients with CF, indicating that extensive removal of infected tissue influences the pathogenic Phosphoprotein phosphatase process of autoantibody production. The results shown herein are in favour of applying EIGSS in selected patients with CF and for using BPI-ANCA as a surrogate marker for guiding further therapeutic interventions. The paranasal sinuses in patients with cystic fibrosis (CF) are often colonized with CF-lung pathogens, especially Pseudomonas aeruginosa [1, 2]. Bacteria from the sinuses can be aspirated to the lower airways and thereby initiate or maintain deleterious lung infections [3]. Antineutrophil cytoplasm autoantibodies (ANCA) directed against bactericidal/permeability-increasing protein (BPI) are frequently seen in patients with CF [4], especially in those with severe lung damage [5, 6]. IgG BPI-ANCA is common and occur in approximately 70% of patients with CF, whereas IgA BPI-ANCA is found in about 35% [7]. There is a strong association between BPI-ANCA and lung infection by P. aeruginosa, and BPI-ANCA levels are significantly correlated with the severity of lung damage [5, 8].

Digested organs were

further stained with biotinylated-an

Digested organs were

further stained with biotinylated-anti-4–1BBL ex vivo, followed by Streptavidin-PE or Streptavidin-allophycocyanin amplification. Biotinylated anti-4–1BBL-treated 4–1BBL-deficient mice were used as a negative staining control for analysis of 4–1BBL expression. The samples were analyzed using FACScalibur, FACSCanto, or LSR II (BD Biosciences) with Cell-Quest or FACSDiva acquisition software. Data analysis was done using FlowJo software (TreeStar Inc., Ashland, OR, USA). Marrow was flushed from the femurs and tibias of 10 C57BL/6 mice and digested for 45 min at 37°C with 0.2 mg/mL Collagenase P (Roche) and 0.2 mg/mL DNase I (Sigma). The single cells were seeded in Petri dishes at a density CTLA-4 inhibitor of 1–2 × 106 cells/cm2. One day later, nonadherent cells were washed away and the remaining adherent cells were expanded in medium (see Generation of memory T cells in vitro) for up to 25 days. Half of the medium was replaced with fresh medium once a week. On day 25, adherent cells were removed by trypsinization, stained with anti-CD45.2 (ebioscience), anti-VCAM-1 (ebioscience),

biotinylated anti-4–1BBL (19H3), and secondary streptavidin, and assessed by flow cytometry. In addition, CD45-negative cells were sorted into the VCAM-1+ and VCAM-1− population. Yields of CD45−VCAM-1+ cells were approximately 500,000 in all three experiments, whereas yields of CD45−VCAM-1− cells ranged 13,000- 165,000 cells. Total RNA was extracted and purified using RNeasy Micro kit (Qiagen). Random Selleckchem AZD1208 hexamer primers and purified RNA were used for the reverse transcription reaction (Invitrogen). PCR of the cDNA was done using following primers: 4–1BBL forward: 5′-CTT GAT GTG GAG GAT ACC-3′, 4–1BBL reverse: 5′-GCT TGG

CGA ACA CAG GAG-3′, CCL19 forward: 5′-GCC TCA GAT TAT CTG CCA T-3′, CCL19 reverse: 5′-AGA CAC AGG GCT CCT TCT GGT-3′, IL-7 forward: 5′-TCC TCC ACT GAT CCT TGT TC-3′, IL-7 reverse: 5′-TTG TGT GCC TTG TGA TAC TG-3′, CXCL12 forward: 5′-GTC CTC TTG CTG TCC AGC TC-3′, CXCL12 reverse: 5′-TAA TTT CGG GTC AAT GCA CA-3′, actin forward: 5′-GGG AAT GGG TCA GAA GGA-3′, actin reverse: 5′-AAG AAG GAA GGC TGG AAA-3′, GAPDH forward: 5′-AAC TTT GGC ATT GTG GAA GG-3′, and GAPDH reverse: 5′-GGA ID-8 GAC AAC CTG GTC CTC AG-3′. CD8+ memory+ T cells were generated in vitro from OT-I DsRed splenocytes as described [29]. A total of 6 × 106 cells were adoptively transferred into C57BL/6 mice. One day later, femurs were harvested, fixed for 4 h in 4% paraformaldehyde at 4°C, dehydrated in 10, 20, and 30% sucrose solution for 24 h, respectively, and frozen in SCEM embedding medium (Section-Lab Co. Ltd., Yokohama, Japan). Bone cryosections (7 μm) were prepared using Kawamoto’s Film Method [51]. Sections were stained with the following Ab from eBioscience if not indicated otherwise: VCAM-1 (429), CD31 (MEC13.

Characteristic PML lesions have been described as large, subcorti

Characteristic PML lesions have been described as large, subcortical, grey-matter-sparing lesions appearing hyperintense on T2 and fluid-attenuated inversion recovery and hypointense

on T1 scans; contrast enhancement may occur [47]. The anti-CD52 mAb alemtuzumab (Lemtrada®) has been shown to be highly effective and is approved for active relapsing MS in Europe [10-12, 69]. Disease activity is defined as clinical or radiological deterioration [70]. Mechanisms of action include depletion of CD52-expressing T/B lymphocytes, natural killer (NK) cells, dendritic cells and monocytes/macrophages with skewed repopulation leading to a reprogramming of the immune repertoire [71, 72]. Already in earlier studies, patients especially with an early relapsing disease course appeared

to benefit most from alemtuzumab check details treatment, leading to the concept of a therapeutic window relatively early during the disease, when highly active immunotherapy may exert most profound effects [72]. This was reflected in the inclusion criteria for the pivotal Phase III trials Sorafenib order CARE-MS I and II (Comparison of Alemtuzumab and Rebif® Efficacy in Multiple Sclerosis, Studies One and Two). CARE-MS I included active relapsing, therapy-naive MS patients, whereas CARE-MS II focused on relapsing MS refractory to first-line therapy [10, 12]. Especially in terms of disease progression, the latter patient group appeared to benefit most. Whereas current EMA approval is relatively broad [70], careful patient selection

is mandatory, as SADRs have been reported and thorough adherence to safety assessments is necessary. This is stressed by long-term data from the Phase II trial CAMMS223, with one additional SADR (Goodpasture syndrome), but also sustained reduction of disability accumulation and relapse rates compared to active comparator [73], revealing the dilemma of long-lasting efficacy versus potential SADRs. Alemtuzumab is applied intravenously with a first treatment cycle of 12 mg over 5 days, followed by a second therapy cycle over 3 days after 12 months [10, 12, 69]. Further cycles are not intended, but the question of when and how to continue DMD treatment after two cycles is unanswered. There is no class I evidence for different treatment protocols in this indication. During and for 1 month after treatment, acyclovir (200 mg twice daily) has to be administered prophylactically. SSR128129E Therapy surveillance with large treatment intervals, but necessarily close safety monitoring, will be a challenge in clinical practice [74] and emphasizes even more the importance of patient education, counselling and informed consent to assure adherence to safety measures. These include differential blood count, serum creatinine and urine analysis before first administration and monthly afterwards; regular testing of thyroid stimulating hormone (TSH) levels has to be performed before treatment initiation and every 3 months up to 4 years after the last administration [70].

This

This AT9283 supplier would manifest as an increase in the relative proportion of antibodies directed against protective, rather than nonprotective epitopes as

a consequence of more efficient presentation to the MHC class II pathway. The results of the present study are highly encouraging and confirm the feasibility of developing a DNA-based vaccine approach that is capable of eliciting protective immune responses against anthrax and plague. Multi-agent DNA vaccines targeting other dissimilar pathogens, notably viruses and bacteria, are already in development and show great promise (Riemenschneider et al., 2003). One of the challenges facing researchers seeking to develop multivalent vaccines is the need to design formulations

that ensure that the development of the responses to the individual antigens does not interfere with each other (Sedegah et al., 2004; Wang et al., 2007; Shen et al., 2009). Fortunately, this was not observed in the antibody titers to the fusion vaccines; however, survival with the phV-LFn/phPA combination was slightly reduced by one animal (17%) when phLFn-F1 was included. This may reflect competition between the endogenously produced fusion proteins for the same binding site on PA following its expression and binding to the cell surface. Studies are currently in progress to characterize Selleck Wnt inhibitor the basis of the immune enhancing effect observed during this study and to determine whether efficacy against plague can be enhanced as a consequence of codon modification or altering the DNA vaccine composition/formulation. All animal

studies were carried out in strict accordance with the Animals (Scientific Procedures) Act 1986. This work was financially supported by contract #0000106993 between the U.S. Naval Medical Research Center, the Henry M. Org 27569 Jackson Foundation for the Advancement of Military medicine, and Dstl. The authors wish to thank G.K. Paterson, A. Gates, A. Stagg, and S. Perkins for critical technical input. The views expressed in this article are those of the authors and do not necessarily reflect the official policy or position of the Department of the Navy, the Department of Defense, nor the U.S. Government. A.K.-M. is an employee of the U.S. Government. This work was prepared as a part of her official duties. Title 17 U.S.C. §105 provides that Copyright protection under this title is not available for any work of the United States Government.’ Title 17 U.S.C §101 defines a U.S. Government work as work prepared by a military service member of employee of the U.S. Government as part of that person’s official duties. “
“Symptoms of diseases such as rheumatoid arthritis, which is T helper 1 (Th1) dependent, and asthma, which is T helper 2 (Th2) dependent, are influenced by diurnal rhythms and natural regulatory T cells (nTreg).

The data showed consistency with a recent report suggesting the e

The data showed consistency with a recent report suggesting the expression of Il10 mRNA in CD19+ B cells of draining LN of susceptible mice at the first day post-inoculation, and it was shown that B cells play as a source of IL-10 which influences the susceptibility of BALB/c

mice to L. major infection [30]. At the late stage of the infection, augmented expression of this cytokine was documented at W3 and then tended to gradually decrease at W5 and W8 post-infection. DE5 strain showed the highest level of expression at W3 post-infection. It seems that IL-10 along with IL-4 cytokine is responsible for the susceptibility of the BALB/c mice to L. major infection, as suggested before [31]. Hence, our results showed that the contribution of selleck screening library DA39 strain in eliciting Il10 mRNA expression is lower than most strains at 16 h and during the late stage of infection. Taken together, the results of this study show that different strains of L. major display different virulence and induce different patterns of cytokine expression in BALB/c mice. While DA39 strain induced the lowest parasite load, high-level expression of Th1-related cytokines mRNA LY2835219 and higher Ifng/Il4 mRNA ratio in LN of BALB/c mice, the SH25 strain elicited the highest number

of viable parasite in LN of the infected mice and a lower level of Ifng/Il4 mRNA ratio than DA39 strain at 40 h and 8 weeks post-infection. Interestingly, DA39 strain has failed to induce higher expressions of both Il4 and Il10 mRNA, especially at the late stage of the infection. It is noteworthy that in our previous study, similar results in the parasite burden and the generation of IFN-γ induced by DA39 strain were reported at 4 weeks post-infection, however at that study, we reported

higher levels of IFN- produced by DE5 strain than DA39 at W8 post-infection [14]. The reason for this discrepancy may be attributed to the methods used for the cytokine evaluation. It might be considered that the expression of the cytokines mRNA by real-time PCR seems to be a more precise method than assessment of the cytokine in lymphocyte culture. Moreover, the Glutathione peroxidase present study was repeated for three times, and the third experiment results were reported as representative. Therefore, DA39 strain might be considered as an ideal strain for the vaccine studies. In conclusion, our results showed variable parasite loads and different expressions of cytokine mRNA in LN of mice infected with the four strains of L. major. Amongst the four strains isolated from the four endemic areas of Iran and analysed by SSCP, DA39 strain induced lower load of parasites in LN of the inoculated BALB/c mice. Moreover, this strain elicited higher expressions of Ifng and Il12 mRNA and lower expressions of Il4 and Il10 mRNA in draining LN of the infected BALB/c mice at early and late stages post-infection.

L and Y M and a postdoctoral grant from ‘Stichting tegen Kanker

L. and Y.M. and a postdoctoral grant from ‘Stichting tegen Kanker’ to J.A.V.G. The authors declare no conflict of interest. Figure  S1 Claudin-1, claudin-2 and claudin-11 proteins are undetectable in IL-4 or TGF-β stimulated BALB/c thio-PEM. BALB/c thio-PEM were left untreated Tamoxifen clinical trial (N) or were treated for 24 h with IL-4 or TGF-β, after which cell lysates were prepared for Western blot. Cell lysates were also prepared from total mouse brain, liver, kidney and spleen tissue. Table  S1 Basal gene expression levels (DCT ± SEM) in unstimulated naive macrophages. “
“Aicardi–Goutières

syndrome (AGS) is a genetically determined disorder, affecting most particularly the brain and the skin, characterized by the inappropriate induction of a type I interferon-mediated immune response. In most, but not all, cases the condition is severe, with a high associated morbidity and mortality. A number of important recent advances have helped to elucidate the biology of the AGS-related proteins, thus providing considerable insight into disease pathology. In this study, we outline the clinical phenotype of AGS, paying particular attention to factors relevant to therapeutic intervention. We then discuss the pathogenesis of AGS from a molecular

and cell biology perspective. Finally, we suggest possible treatment strategies in light of these emerging click here insights. Other Articles published in this series Mouse models for Aicardi–Goutières syndrome provide clues to the molecular pathogenesis of systemic autoimmunity.

Clinical and Experimental Immunology 2014, 175: 9–16. Aicardi–Goutières syndrome: a model disease for systemic autoimmunity Clinical and Experimental Immunology 2014, 175: 17–24. We have previously published a description of the genotype–phenotype correlation in 121 patients with Aicardi–Goutières syndrome (AGS) [1]. Based on that work, and an ongoing exercise to assimilate clinical and laboratory data from >250 cases (http://www.nimbl.eu/ni/Home), the natural history of AGS is becoming clearer. In a significant minority of patients with AGS, problems are recognized Cobimetinib at birth, i.e. the disease process begins in utero. Over time, severe neurological dysfunction manifests as progressive microcephaly, spasticity, psychomotor retardation and, in approximately 35% of cases, death in early childhood. Typical clinico-radiological features include intracranial calcification, white matter changes and raised numbers of white cells in the cerebrospinal fluid (CSF). To a remarkable degree this form of the disease, seen most consistently in association with mutations in TREX1, RNASEH2A and RNASEH2C, mimics the sequelae of congenital, transplacentally acquired infection (hence the tag: ‘pseudo-TORCH’ syndrome – Toxoplasmosis, Rubella, Cytomegalovirus and Herpes) [2]. More frequently, a later-onset presentation of AGS is seen, occurring in some cases after several months of normal development [3, 4].

[16] reported unaltered spontaneous apoptosis rates Since the tw

[16] reported unaltered spontaneous apoptosis rates. Since the two mouse models were designed in the very same way, these different observations may be due to different integration sites of the transgenes in the genome. Of note, we did not detect any differences in spontaneous apoptosis of thymocytes between WT and vavFLIPR mice. Gene-targeting studies revealed that c-FLIP is crucial for efficient T-cell development [27, Cytoskeletal Signaling inhibitor 28]. On the other hand, transgenic mice expressing c-FLIPL in a T-cell-specific manner exhibited disturbed T-cell development, reduced positive selection

and, at least in the BALB/c background, developed autoimmunity [29, 30]. Thus, T-cell development appears to require a balanced expression of c-FLIP. In contrast to c-FLIPL transgenic mice, we did not observe alterations in T-cell cellularity, frequencies of the main T-cell subsets in thymus and peripheral lymphoid organs, and in the activation status of

CD4+ and CD8+ T cells in vavFLIPR mice. Therefore, we conclude that c-FLIPR does not have a functional role in T-cell development. Lpr and gld mice, which have natural occurring mutations in the CD95 and CD95L genes, respectively, exhibit lymphoproliferative disease and autoimmunity [31]. It was therefore expected that transgenic overexpression of c-FLIP proteins results in autoimmune disease find more as well. However, T-cell-specific expression of murine c-FLIPL or human c-FLIPS did not recapitulate the lpr/gld phenotype [15, 16, 26]. Similarly, we did not observe lymphoproliferation at 3–5 months of age in vavFLIPR mice. Rather, T-cell development and distribution of lymphocyte subsets appeared normal in vavFLIPR mice. In contrast to the cellular FLIP proteins, the situation is more complex for

viral FLIP proteins. For instance, expression of MC159, a vFLIP from the human Molluscum contagiosum virus under the control of a CD2 enhancer cassette, did not result in lymphoproliferation [32]. On the other hand, an lpr/gld-like phenotype was observed when MC159 was placed ifenprodil under the control of the ubiquitous MHC class I H2Kb promoter [33]. Therefore, inhibition of death receptor-mediated apoptosis in non-T cells seems to be crucial for the development of autoimmunity, which is consistent with the observation that lack of CD95 expression in DC results in systemic autoimmunity [34]. To analyze the in vivo effect of c-FLIPR overexpression, we challenged the vavFLIPR mice and their WT littermates with L. monocytogenes. Infection with this gram-positive intracellular bacterium is a well-established model for analysis of the adaptive immune response [24]. Moreover, T cells are known to be required for the resolution of L. monocytogenes infection and protective immunity [35]. Strikingly, the vavFLIPR mice were more efficient in clearing the bacterial load and showed less liver necrosis and less caspase-3 activation.

This observation strongly argued in favour of a general regulatio

This observation strongly argued in favour of a general regulation of immune response by corticoid hormones during H. polygyrus infection [12, 28]. In the present study, we identified that H. polygyrus

products are potent to inhibit apoptosis provoked by DEX in MLN cell populations. The most sensitive subpopulation was CD4+CD25hi cells. Significantly, more CD4+CD25hi cells than other subpopulation of T cells underwent apoptosis 12 days after infection; it might be that activated via TCR, CD4+CD25hi cells expressed a high level of glucocorticoid-induced TNF receptor, Decitabine GITR and therefore this subpopulation was more sensitive to glucocorticoid-induced apoptosis, which was previously reported [29, 30]. The inhibition of apoptosis induced via TCR receptor in MLN cells exposed to H. polygyrus antigen in vitro is confirmed by the elevated expression of FLIP, which is an inhibitor of death receptor-mediated apoptosis via caspase cascade. FLIP is expressed click here during the early stage of T-cell activation, but disappears when T cells become susceptible to Fas ligand-mediated apoptosis [31, 32]. High expression of FLIP protein was present both in naïve and restimulated cells and was distinctly regulated by H. polygyrus antigenic fractions. Heligmosomoides polygyrus infection

and the nematode protein fractions activated FLIP in MLN cells. The studies of different populations of lymphocytes revealed significant differences in the percentage of apoptotic cells between control and infected mice. The antigenic fractions added to the culture supported survival of cells preferentially from infected mice. As the level of apoptosis was different and FLIP expression

did not correlate with the infection, it is likely that FLIP would not be considered as a specific marker of inhibited apoptosis during H. polygyrus infection. Naïve cells which expressed FLIP were also sensitive to DEX-induced apoptosis in spite of exposure to H. polygyrus antigens in cell culture. It seems that signals other than only FLIP were required to keep cells alive. DEX induces apoptosis via the intrinsic mitochondrial pathway [33]; therefore, H. polygyrus related factors were probably able to induce those signals which produce Bcl-2, but only after restimulation. This was also reflected in the higher percentage of Bcl-2-positive CD4+ Thiamet G T cells, which were evoked by factors present in all examined antigen fractions. The nematode infection induces expansion of CD8+ T regulatory cells [34]. We indicated that survival of CD8+ T-cell population was regulated differently than of CD4+ T cells; both infection and restimulation with H. polygyrus antigen strongly reduced the percentage of Bcl-2-positive cells among T-cell subpopulations [12]. The percentage of CD4+ T cells which expressed Bcl-2 protein increased but the percentage of CD8+ T cells was strongly reduced. This might suggest that H.

In addition, immunostimulants such as CpG DNA inhibit DC apoptosi

In addition, immunostimulants such as CpG DNA inhibit DC apoptosis 18, whereas the deficiency of pro-apoptotic Bim protein in DC results in autoimmunity 19. Immature DC have the ability to acquire protein complexes or soluble antigen using many different pathways such as macropinocytosis, endocytosis and even through ingestion of entire cells. Despite the importance of DC apoptosis in the immune response, studies have not investigated the

effects of DC death on viable DC. In this study, we show that viable Sorafenib price immature DC have the ability to uptake apoptotic DC. The uptake of apoptotic DC or necrotic DC is recognized as an immunologically null event. However, it is the uptake of apoptotic DC that suppresses subsequent maturation of viable DC in response to LPS and results in upregulation of TGF-β2 and preferential secretion of TGF-β1, which mediates induction of naïve T cells into Foxp3+ Treg. In contrast, the uptake of apoptotic splenocytes by viable immature DC does not result in TGF-β1 secretion, nor does it result in induction of Foxp3+ Treg. Therefore, it is likely the uptake of apoptotic DC by viable DC that provides a potential to induce Foxp3+ Treg. Bone-marrow-derived DC were treated with UV light, and apoptosis

induction was assessed at 1 and 6 h after UV treatment. Prior to UV treatment, cells were mostly positive for Hoechst 3342 (a cell permeant DNA-binding stain, blue) with very few cells being PLX-4720 ic50 positive for annexin V (a phosphatidylserine-binding protein, green), indicative of live DC. One hour after UV treatment, majority of the cells

were positive for both annexin V and Hoechst 3342, with very few cells positive for ethidium homodimer (EH) (a nuclei probe, impermeant to live RVX-208 or apoptotic cells, red) (Fig. 1A). In these cells, there was translocation of phosphatidylserine on the membrane as indicated by positive annexin V staining, but the membrane integrity was still maintained, as they were mostly negative for EH stain; hence, they can be classified as apoptotic cells. In contrast, 6 h after UV treatment, there was a pronounced increase in EH positive cells, indicating that the membrane integrity was compromised. However, these cells were also positive for annexin V (Fig. 1A). Therefore, these cells can be classified as late apoptotic cells. In order to further confirm apoptosis in a quantitative manner, 1 or 6 h after UV treatment, DC were stained with annexin V and propidium iodide (PI), and apoptosis was assessed via FACS analysis. Prior to UV treatment, approximately 10% of DC were annexin V+PI–, whereas 1 h after UV treatment approximately 45% of DC were annexin V+PI–, indicative of apoptotic cells and confirming our above findings (Fig. 1B). At 6 h post-UV treatment, approximately 80% of cells were annexin V+PI+, indicating that these cells were in late apoptosis (Fig. 1B).