One wonders what actually determines the “helper dependence” of a

One wonders what actually determines the “helper dependence” of an immunogenic virus, and whether the experimentally observed differences might be related to intrinsic features of the various pathogens or perhaps the dose at which they are offered as immunogens?

After all, immature DC have been shown to acquire CD8+ CTL priming capacity by both T-helper-independent or -dependent stimuli 8. It seems not unreasonable to suppose that T help is required under limiting doses AP24534 cost of danger signals (TLR ligands, NOD ligands and type I IFN), in which case CD40L signaling by CD4+ helper cells and resulting cognate events are required for appropriate DC activation followed by CD8 (re)activation. One intriguing aspect of the Baker et al.1 study is their finding that CD8+ T cells lacking the IL-21 receptor have a significant induction of TRAIL expression in a manner similar to “helpless” CD8+ T cells primed in the absence of CD4+ T cells 2. The most

likely source of the IL-21 in this scenario is the CD4+ T cell, although NKT cells have not been excluded. This leads to the idea that one previously unanticipated role of T help is to control secondary expansion via regulation of TRAIL expression in CD8+ T cells. This raises a number of interesting questions regarding the time and place of cytokine signals in the provision PD0332991 research buy of T help. For instance, when is IL-21 signaling important for CD8+ T cells? How might this fit with the finding of Bevan’s group that CD8+ T cells must Tryptophan synthase receive IL-2 signals during the first 6 days of priming in order to become capable of secondary expansion 9? Must CD4+ T cells produce both IL-2 and IL-21

or might these two γ chain cytokines serve a redundant function? If both are required, might they be produced simultaneously or sequentially? How does the requirement for these cytokines correspond with the apparently conditional need for cognate interactions among CD4+ T cells, DC and CD8+ T cells? CD8+ T-cell effector and memory responses need to be tightly controlled for several reasons, including rapid induction of robust killer cell function when needed, rapid recall in case of dangerous reinfections and avoidance of massive auto-destruction by runaway auto-reactive CTL. Control of CD8+ T cells is mediated by a variety of intricate cognate interactions between CD4+ helper cells, DC and CD8+ cell precursors. These interactions determine the quality of the DC activation and subsequent CD8+ CTL precursor activation. Crucial events are CD40 ligand (CD154) upregulation on CD4+ helper cells, followed by DC activation through CD40 ligand triggering of CD40 on DC 10, 11.

Similar events may be initiated in T cells by ICs and complement

Similar events may be initiated in T cells by ICs and complement activation in autoimmune disorders. Syk has been a target for therapeutic intervention for autoimmune diseases. Syk-mediated signalling contributes to the altered T cell signalling [31]. In this report, we demonstrate that the FcγRIIIA/B receptor engagement by ICs on CD4+

T cells leads to the recruitment of the signalling subunit, the FcRγ chain, thus resulting in Syk activation. The Akt inhibitor presence of soluble TCC enhances this signalling event. TCC in fluid phase by associating with vitronectin (S protein) becomes cytolytically inactive and is regarded as irrelevant. However, recent reports have shown that TCC induces functional activities such as kinin-dependent vascular leakage, activation of endothelial cells and induction of osteoprotegerin [16,32,33]. Vitronectin facilitates the cellular adhesion of soluble TCC, providing a mechanism to trigger cellular responses [34]. Previously, we have shown elevated levels of vitronectin associated with membrane attack complex (MAC) in lupus nephritis patients [23]. Our results point to a synergistic

role for TCC in IC-mediated Syk activation in CD4+ T cells. Such synergistic action of ICs and MAC in chemokine secretion during lung tissue injury has also been reported previously [35]. Binding of AHG (Fig. 1) and ICs purified from SLE Carnitine dehydrogenase to the peripheral CD4+ T cells establishes the interaction and a possible role of ICs in T cell responses. Previously, activation-dependent selleck chemicals llc expression of FcγRII and FcγRIII receptors in the human T lymphocyte subpopulation has been observed [36]. This study showed a four- to 10-fold increase in the FcγRIII+ CD8+ T cell population in response to phytohaemagglutinin (PHA) treatment on day 3 post-stimulation [36]. Our results also point to a similar phenomenon, where FcγRIII+CD4+

T cells expanded in vitro using anti-CD3 and CD28, a total of more than 40% cells stained for FcγRIIIA/B in comparison to 10% directly from the PBMC. To explore whether ICs can influence the T cell physiology, we investigated the role of these complexes in Syk activation. Syk is a homologue of non-receptor tyrosine kinase ZAP-70. Syk is activated by FcRγ chain upon ITAM phosphorylation. Syk is expressed widely in both immune and non-immune cells [37,38]. Both DAP-12 and FcγR associate with Syk and mediate β-2 integrin signalling in neutrophils and macrophages [39]. Syk phosphorylation also occurs upon engagement of pathogen recognition receptors such as FcγR, CR3 and Dectin-1 [1]. Accumulating evidence points to Syk expression in subsets of T lymphocytes such as thymocytes, naive αβ T cells and intraepithelial γδ T cells, but not in proliferating and mature T cells [31,40].

Patients were informed about the aim of the study and gave their

Patients were informed about the aim of the study and gave their full consent. The study was approved by the Ethical Committee of

Department of Pediatrics, University Federico II, Naples. The serum level of endomysium (EMA) and tissue transglutaminase (anti-tTG) antibodies [immunoglogbulin (Ig)A] was measured immediately before both gluten challenges started (day 0). EMA were detected by indirect immunofluorescence on frozen sections of human umbilical cord and anti-tTG using the enzyme-linked immunosorbent assay (ELISA) technique with a commercial kit (Eu tTg IgA; Eurospital, Trieste, Italy). Results were interpreted according to the manufacturer’s instructions: negative <9 U/ml, weak positive in the range 9–16 U/ml, check details positive >16 U/ml. Patients ate 200 g of wheat bread or cookies daily for 3 days, corresponding to about 12 g of gluten per day (first challenge). After a wash-out of 3–10 months on a strict gluten-free diet, 13 of 14 coeliacs consumed wheat for an additional 3 days (second challenge). At the time of the first gluten challenge, 11 patients were seronegative for EMA or anti-tTG and three had low antibody titres. Two patients complained about abdominal pain on the first day of the challenge, but they did not stop the gluten intake. The remaining patients reported no symptoms. A commercial wheat flour was used for baking the bread and

cookies. Gliadin was extracted according to Wieser selleck products et al. [20] and digested enzymatically with pepsin and trypsin, as described previously [21]. The 33-mer (α-gliadin 57–89) peptide was synthesized by solid-phase automated flow, as described elsewhere [2]. Both PT-gliadin (indicated hereafter as gliadin) and peptides were deamidated with guinea pig tTG, as reported elsewhere [2]. Venous blood (15–20 ml) was collected in a heparizined syringe before (day Montelukast Sodium 0) and 6 days after (day 6) the gluten challenge. Peripheral blood mononuclear cells (PBMCs) were isolated by Ficoll-Paque density centrifugation. PBMCs were analysed immediately for antigen recognition by IFN-γ ELISPOT assay, as described previously [22]. Briefly, 4 × 105 PBMCs were seeded in 200 µl

of complete medium X-Vivo15 supplemented with 5% heat-inactivated AB pooled human serum, 1% antibiotics (100 U/ml penicillin and 100 µg/ml streptomycin) and 1% L-glutamine (2 mM) (all provided by BioWhittaker, Verviers, Belgium) in duplicate in 96-well plates (Millipore, Bedford, MA, USA) coated with purified anti-human IFN-γ antibody (MabTech, Nacka Strand, Sweden). Gliadin, either deamidated or native, was tested at 50 µg/ml and 33-mer peptide at 30 µg/ml (7·7 µM). Cells were incubated for 36–40 h with biotinylated anti-human IFN-γ antibody (MabTech) followed by incubation with streptavidin horseradish peroxidase (HRP) (BD-Pharmingen, San Diego, CA, USA). Spot-forming cells (SFC) were counted by an immunospot analyser (A.EL.

After 3 days, HSCs were isolated from the bone marrow After 10 d

After 3 days, HSCs were isolated from the bone marrow. After 10 days in culture, 1×105 cells of two different HSC populations were injected into Rag-2/γC−/− mice expressing either H-2Kd or H-2Kb. Mice were analyzed 4–5 wk after HSC transfer. Animal experiments were done in compliance with the guidelines of German law and the Max-Planck-Institute of www.selleckchem.com/products/AZD1152-HQPA.html Immunobiology and Epigenetics. HSCs were grown in Iscove’s medium (Biochrom) supplemented

with 2% of heat inactivated FCS (PAN Biotech), 10 mM L-glutamine, 100 U/mL penicillin, 100 U/mL streptomycin (GIBCO), 50 mM 2-mercaptoethanol, 0.03% primatone (Sigma-Aldrich), 4.2 mg/mL insulin (Sigma-Aldrich), IL-6, IL-3 and c-kit-ligand. The expression of H-2d and H-2b was determined by flow cytometry using the specific monoclonal antibodies H-2Dd-PE and H-2Kb-FITC

(BD). Cells were stained with anti-B220/CD45R-PerCP (RA3-6B2, BD), anti-CD43-PE (S7, BD), anti-CD19-PE/-PerCP (1D3, BD), anti-CD21-APC (7G6, BD), anti-CD23-PE/biotin (B3B4, BD/PharMingen), anti-IgM-Cy5 (Jackson Immunoresearch) and anti-idiotype 54.1 (kindly provided TSA HDAC research buy by D. Nemazee). Flow cytometric analysis was performed with FACS-Calibur (BD). Statistical analysis was performed with the GraphPad Prism 4 software using Student’s t-test as the statistical hypothesis test. The authors thank U. Stauffer, N. Joswig and C. Johner for mouse work and further assistance. They thank E. Hobeika for the mb1-lox-GFP mice, P. Nielsen, D. Nemazee and M. Reth for critical reading of the manuscript. This work was supported by the Deutsche Forschungsgemeinschaft (SFB620 and SFB746). Conflict of interest: The authors declare no financial or commercial conflict of interest. Detailed facts of importance to specialist readers are published as ”Supporting Information”.

Such documents are peer-reviewed, but not copy-edited or typeset. They are made available as submitted by the authors. “
“Two outbreaks of Streptococcus suis ST7 occurred in humans in 1998 and 2005 in China. PFGE of either chromosome restriction fragments found all ST7 isolates to be indistinguishable. Due to the genetic homogeneity of ST7 isolates, development of a rapid sub-typing method with high discriminatory power for ST7 isolates is required. In this study, a novel method, MLVA, was developed to type S. suis serotype 2 strains. Further, this method was used to analyze outbreak-associated ST7 strains in China. A total of 144 ST7 S. suis isolates were sub-typed into 34 MLVA types. Among these, eight isolates from the 1998 outbreak were sub-typed into five MLVA types, of which four MLVA types were also detected in Sichuan in 2005. These data indicate that the pathogens responsible for the two outbreaks had the same origin. In addition, some observations also provided molecular evidence for the transmission route, possibly indicating that the MLVA method has usefulness in epidemiology. The developed MLVA scheme for S.

Undoubtedly, the laboratory mouse has proven to be an invaluable

Undoubtedly, the laboratory mouse has proven to be an invaluable model for biological research and most of what we know today about mammalian biology is derived from research carried out with Mus musculus. Nonetheless, to reject other animal models is to ignore the

need to address evolutionary divergence among mammals by studying biology across an array of genotypes. Moreover, the opportunity to exploit unique biological models or intriguing insights can be squandered. Clarity about the nature of immunologic tolerance was developed because Owen and Medawar capitalized on the unique properties of the placental vasculature of twin calves. Rowson’s frustrations with uterine infections in embryo transfer recipients gave impetus to his fruitful ABT-199 in vivo studies that established progesterone as a key hormone regulating uterine immunity. The papers in this special issue of the American Journal of Reproductive Immunology highlight additional examples whereby farm animals are being used to develop

concepts pertinent to a wide range of mammalian species. Domestic farm animals are not the only mammalian species that can make useful research models, of course, but they offer advantages of availability, ease of handling, cost, and a well-described biology and husbandry. When Medawar was struck with the idea of using the calf in his research, he RG7204 order turned to colleagues at the Animal Breeding and Genetics Research Organization in Edinburgh. Today, unfortunately, the infrastructure for conducting farm animal research is eroding.21,22 For example,

the number of scientist years working in animal production or protection in the United States declined 22% from 1985 to 2006 and doctorates awarded in the animal sciences in the United States declined by 30% from 1985 to 2004. An increase in selleck products investments in basic research using farm animals will have a positive impact not only on agricultural productivity but on understanding mammalian biology and enhancing human health. During the initial preparation for this paper, I was fortunate enough to attend the celebrations surrounding the 100th Anniversary of the Dept. of Genetics at the University of Florida. In the course of the event, I heard details of the contributions of Ray Owen to the idea of immunologic tolerance that I was unaware of previously. Medawar had acknowledged his debt to Owen in his Nobel Lecture but, until I heard the details in Madison, I knew little about Owen or his work. I acknowledge Millard Susman, James Crow and Ray Owen for sharing images and information about this important time in reproductive immunology. “
“This study investigated whether angiotensin II type 1 receptor agonistic autoantibodies (AT1-AAs) mediate the increased release of soluble endoglin (sEng) in women with preeclampsia. Serum samples were obtained from women with normal pregnancies or with preeclampsia.

The purpose of our study was to analyse the prevalence of Malasse

The purpose of our study was to analyse the prevalence of Malassezia species in lesional skin of SD, and to assess the distribution of the species according to severity of the disease and cellular immune status of the patients. Forty SD patients with scalp involvement were included in the study. The samples were obtained by scraping the skin surface of the scalp selleck kinase inhibitor and then incubated on Sabouraud dextrose agar and modified Dixon agar. The yeasts isolated were identified by their morphological and physiological properties according

to the method of Guillot et al. In addition, we performed two-colour flow cytometry analysis to investigate the lymphocyte subpopulations in the peripheral blood. The most commonly isolated species was Malassezia restricta (27.5%), followed by Malassezia globosa (17.5%) and Malassezia see more slooffiae (15%). We demonstrated low helper/suppressor ratios in 70% patients, because of an increase in the suppressor T-cell population, suggesting an impaired cellular immunity. However, we found no significant difference

in the distribution of isolated Malassezia species according to the severity of the scalp involvement and changes in the peripheral blood lymphocyte subpopulations. “
“We report Schizophyllum commune as the aetiological agent of one case each of allergic broncho-pulmonary mycosis (ABPM) and pulmonary fungal nearly ball, and present a literature review. The fungus was characterised by

clamp connections, hyphal spicules, and formation of basidiocarps with basidiospores. The phenotypic identification was confirmed by sequencing of the ITS region. To-date, ABPM and pulmonary fungal ball to S. commune have been reported exclusively from Japan and North America respectively. Of the 71 globally reported cases due to S. commune, 45 (63%) were bronchopulmonary, 22 (31%) sinusitis and 4 extrapulmonary. Taken together, cases of bronchopulmonary disease and sinusitis numbered 67 (94%), indicating the respiratory tract as the primary target of disease. Concerning the country-wise distribution, Japan topped the list with 33 cases (46%), followed by Iran – 7 cases (10%), U.S.A. – 6 cases (9%), and a lower prevalence of 1.4–6% for the remaining 12 countries. The preponderance of the disease in Japan may be attributed to its greater awareness vis-à-vis that in other countries rather than to any geographical/climatic factors. We believe that the burden of S. commune-incited disease is currently underestimated, warranting comprehensive prospective studies to determine its prevalence. “
“Triazole and imidazole antifungal agents inhibit metabolism of vincristine, leading to excess vinca alkaloid exposure and severe neurotoxicity.

These results imply that the species of protozoa available for P

These results imply that the species of protozoa available for P. acanthamoebae in the natural environment are limited. Observations from the FISH and TEM analyses support the data obtained from the AIU assays.

The inclusions that formed within P. acanthamoebae following infection of Acanthamoebae were relatively small, when compared with the inclusions which form in epithelial or immune cells infected with pathogenic chlamydiae (25–27). Although the exact reason for selleck chemical this difference is unknown, it is possible that rapid growth and maturation of the bacteria occurred following their uptake into Acanthamoeba. It is well established that formation of inclusions due to infection with pathogenic chlamydiae is seen in a wide variety of mammalian cells regardless of the cell type (28–32). However, there was no evidence of inclusion bodies or growth of P. acanthamoebae in the mammalian cells used in our study. selleck chemicals This result is controversial because previous studies have demonstrated that P. acanthamoebae is able to enter, and multiply within, human pneumocytes, lung fibroblasts and macrophages (19–21). The exact reason for this difference remains unknown, but this contradiction may be associated with

difference in culture conditions or in the traits of the cell lines used. In either case, taken together with the present findings, it is concluded that the host range of P. acanthamoebae is limited, implying that Acanthamoebae is a unique reservoir for the bacteria in nature, and that growth of P. acanthamoebae in phagocytic or non-phagocytic mammalian cells is minimal. Although there one study did show that P. acanthamoebae can induce severe pneumonia in mice (9), it could not be shown whether lung inflammation was caused by stimulation with unknown antigens derived from the bacteria or by bacterial growth in the macrophages or pneumocytes. The P. acanthamoebae Bn9 strain was only used for this

study; other strains were not assessed because of unavailability. Meanwhile, in 17-DMAG (Alvespimycin) HCl this study it was found that Protochlamydia, an environmental strain which is related to Parachlamydia and is a stock collection in the authors’ laboratory, could not grow within mammalian cells as well as Parachlamydia (data not shown), supporting the contention that the host range of P. acanthamoebae is limited. In conclusion, these results indicate that the host range of P. acanthamoebae is limited, and that the AIU assay for quantifying the infective progeny of P. acanthamoebae could be a promising tool for monitoring exact numbers of P. acanthamoebae in host cells, comparable to the inclusion-forming unit assays available for chlamydia such as C. pneumoniae and C. psittaci. The method previously established by the present authors is useful for understanding the dynamics of P. acanthamoebae with respect to potential pathogenic behavior in humans.

We first observed that anti-mCD20 mAb (18B12) efficiently deplete

We first observed that anti-mCD20 mAb (18B12) efficiently depleted B cells in the periphery and spleen and to a lesser extent in the peritoneal XL184 molecular weight cavity for a long time-period, in agreement with previous findings [17]. Baseline serum IgG levels were unaffected, presumably because the majority of antibodies are produced from CD20- plasma cells [11]. However, the outcomes of anti-CD20 mAb-mediated B cell depletion on T cell subsets in the previous studies are controversial. Thus, a slight increase in the percentages of naive CD4+ and CD8+ T cells

(CD44lowCD62Lhigh) and a decrease in memory T cells (CD4+CD44highCD62Llow) were reported in one study [17] but not in another study [8]. Furthermore, expansion of regulatory T cells (Treg) was demonstrated recently in some studies [28,29] but not another study [30] in non-obese diabetic (NOD) mice. In this study we found no change in naive/activated/memory T cell subsets and also in Treg subsets. In the Graves’ mouse model we then showed the excellent prophylactic effect of anti-mCD20 mAb for blocking induction of anti-TSHR antibodies and preventing

hyperthyroidism. This outcome could be expected because anti-mCD20 mAb eliminated antibody-producing B cells almost completely before immunization. However, B cell depletion before immunization also suppressed antigen-specific T cell activation learn more significantly in a T cell recall assay. Previously, suppression of in vitro T cell proliferation and/or proinflammatory cytokine [IFN-γ and interleukin (IL)-17] secretion was reported [22,30], as well as in vivo proliferation of autoreactive T cells in response to endogenous autoantigens by B cell depletion [8]. Thus, elimination of both antigen-presentation and

antibody production by B cells is possibly involved in this highly efficient prophylactic effect. The effect of B Branched chain aminotransferase cell depletion by anti-mCD20 mAb persisted even after the recovery of B cell numbers, as reported previously in diabetes [30]. B cell depletion may be able to ‘reset’ the immune system by breaking the self-perpetuating vicious cycle of autoreactive B cell generation and T cell activation. However, in other cases, continuous B cell depletion was necessary [19]. It is therefore critical to clarify the reason(s) of these differences for optimizing treatment strategies. B cell depletion after the first immunization, when T cells were primed but anti-TSHR antibody production was not observed, was also effective at reducing hyperthyroidism, albeit to a lesser extent than when given before the first immunization.

Moreover, canakinumab significantly reduced the risk of recurrent

Moreover, canakinumab significantly reduced the risk of recurrent flares as compared with triamcinolone acetonide. Thus, neutralization of IL-1β provides rapid and sustained pain relief and reduced the number of recurrent flares compared with steroid use. Despite the availability of several widely used TNF-α-blocking therapies for rheumatoid arthritis and other auto-immune diseases, there is a paucity of reports that blocking TNF-α provides an effective reduction in gout severity. One explanation for the lack of clinical trials of TNF-α blockade

in gout attacks is that the efficacy of TNF-α blockade in refractory gout is less than expected. One study reports a weak selleck chemicals response with rather high doses of infliximab 81. There are also few publications on MSU crystals inducing TNF-α from human and mouse cells unless co-stimulated with endotoxins. Therefore, IL-1β blockade may be used for inducing long-term

remissions in refractory patients and replace glucocorticoids. If IL-1β blockade Selleckchem Bortezomib becomes the standard of care in refractory gout, it would be consistent with the unique role of IL-1β in the pathogenesis of auto-inflammatory diseases. The evidence that IL-1β was toxic for the insulin-producing β-cell begins in 1985 using anti-human IL-1β immunoaffinity chromatography 82. This was a milestone report that advanced the field of “soluble factors” from mononuclear phagocytes playing a pivotal role in the pathogenesis of diabetes. Soon thereafter, recombinant human IL-1β was shown to account

for the death of the β-cell while sparing the α-cell 83. The topic has been acetylcholine reviewed by Mandrup-Poulsen and co-workers, Mandrup-Poulsen being responsible for the original studies 84. Initially, IL-1 was considered to play a pathogenic role primarily in type 1 diabetes, but a role for IL-1β in type 2 diabetes was not appreciated at that time. However, from the studies of Donath et al., IL-1β was implicated in type 2 diabetes, which supported the concept that type 2 diabetes is a chronic inflammatory disease (reviewed in 84). In fact, it was shown that high concentrations of glucose stimulated IL-1β production from the β-cell itself 85 resulting in β-cell death and progressive loss in β cell mass. Relevant to the pathogenesis of type 2 diabetes, glucose-induced IL-1β from the β-cell is enhanced by the presence of free fatty acids. Fundamental to IL-1β-mediated loss of β cell mass is the metabolic upheaval of over-nutrition and obesity and there studies show that the adipocyte in the distant fat stores contributes to the loss of the β-cells 86. The loss of the β cell by IL-1β can also be mediated by oligomers of islet amyloid polypeptide, a protein that forms amyloid deposits in the pancreas during type 2 diabetes, triggering NLRP3 and generating mature IL-1β 87.

Anti-inflammatory agents, such as glucocorticoids and VIP, can di

Anti-inflammatory agents, such as glucocorticoids and VIP, can directly suppress the function of monocytes and macrophages and result in the inhibition of TLR4 ligand-induced TNF-α production.9 In contrast, GPC81–95 inhibits TLR4 ligand-induced TNF-α production by generating CD4+ T cells with anti-inflammatory properties. GPC81–95 stimulates LAP (TGF-β1) expression on only a small

fraction of primary CD4+ T cells (1–2·6%) or Jurkat T cells (3–4%). It is likely that specific receptor(s) are involved in the recognition of the identified peptide and the expression of these receptors may be up-regulated in a small population of primary CD4+ T cells. However, this hypothesis may

not BTK inhibitor explain why only a small population of Jurkat T cells responded to the peptide stimulation. It is possible, but not proven, that up-regulation of LAP (TGF-β1) is confined screening assay to the physiological condition of cells such as a stage of cell division. The fact that only small population of CD3+ CD4+ T cells responded to anti-CD3 antibody and expressed LAP (TGF-β1) supports this notion. Although, the majority of CD4+ T cells express CD3 molecules but only a small population of CD3+ CD4+ T cells responded to anti-CD3 antibody and expressed LAP (TGF-β1). Anti-CD3 antibody is the only known ligand that induces LAP (TGF-β1) expression on CD4+ T pheromone cells and the administration of this antibody suppresses inflammatory conditions in a TGF-β1-dependent manner.3,27 Our data have shown that both GPC81–95 and anti-CD3 antibody induce LAP (TGF-β1) on primary CD4 T cells. It has been suggested that GARP (glycoprotein-A repetitions predominant) is essential for surface expression of

LAP (TGF-β1) on activated regulatory T cells.1 In our hands, no positive cells were detected in resting primary CD4 T cells using the only commercially available anti-GARP antibody (LRRC32 monoclonal antibody; Enzo Life Science, Exeter, UK) (isotype control IgG2b; Enzo Life Science). Using this antibody, no positive cells were detected in GPC81–95 or anti-CD3 antibody-induced LAP expressing primary CD4 T cells (data not shown). Therefore, we are unable to confirm or exclude the possibility that GARP may be expressed on these cells. Further studies are planned to demonstrate whether GPC81–95 can induce LAP (TGF-β1) expression and inhibit inflammation in an in vivo model. Previously self-derived synthetic peptides that exert immunoregulatory effects via induction of TGF-β1 and activation of regulatory T cells have been described. These peptides are derived from a conserved region of the MHC class II molecule and are shown to bind to the MHC and alter T-cell receptor (TCR) –MHC interaction, thereby exerting their inhibitory effect via the TCR.