Cells were left in culture for 4 days in 5% CO2 at 40 °C At day 

Cells were left in culture for 4 days in 5% CO2 at 40 °C. At day 4, EDTA (20 mm) was added to all wells to a final concentration of 2 mm EDTA. The plate was left for 10 min in the CO2 incubator at 40 °C to detach cells from the well. Finally, each sample was mixed by carefully pipetting up and down before transferring

it to FACS tubes. Flow cytometry.  Staining was carried out in tubes by adding 110 μl cell suspension to 90 μl of FACS buffer (0.2% BSA, 0.2% sodium azide, 0.05% normal horse serum in PBS) containing CD4-RPE (Clone CT4) and CD8α-APC (Clone CT8 or Clone 3-298) or CD8α-RPE (Clone EP72 or Clone 3-298). All antibodies were purchased from SouthernBiotech (Birmingham, AL, selleckchem USA). In addition, propidium iodide (Fluka BioChemica, Buchs, Switzerland) was added to exclude dead cells. Cells were incubated at 4 °C for 15 min and then washed once with 2 ml FACS buffer by centrifugation at 295 g for 5 min. All flow cytometry analyses were

performed on a BD FACSCanto™ (BD Biosciences, San Jose, CA, USA) equipped with a 488-nm blue laser and a 633-nm red laser. Using the FACSDiva software, we aimed at collecting a minimum of 10,000 live cells from each sample. Titration of all antibodies was performed prior to the experiment in order to determine the optimal staining concentrations, Midostaurin cost and the multicolour panel was carefully evaluated using fluorescence minus one (FMO) controls [15]. Statistical analysis.  Antigen-specific stimulations were run in triplicates with the exception of the experiment where the effect of anticoagulant and type of serum were tested, as this experiment was run in duplicates. For all optimization steps much and for experiment 1, the mean percentage of proliferated cells ± SE was calculated and shown. The CFSE proliferation data for experiment 2 were tested using standard anovaF-tests on a 5% significance level, with dose and breeding line as the classification variables. Normally, blood is stabilized with heparin, and FBS is used as an additive to growth medium

in cellular stimulation assays. We wanted to test EDTA as a substituent for heparin as anticoagulant in the blood samples and autologous serum from an NDV-vaccinated chicken (CIS) as a substituent for FBS in the cell culture medium used for our recall proliferation assay assessed for both CD4+ and CD8α+ (Fig. 1A) T cells. The strategy for gating on CD4+ and CD8α+ T cells was debris exclusion on the Forward Scatter (FSC) – Side Scatter (SSC) dot plot followed by exclusion of dead cells by PI staining. Out of the live cells, CD4 cells were gated positive at the PE axis and CD8α cells were gated positive at the APC axis in a PE-APC dot plot (Fig. 1A). Finally, the CD4+ and CD8α+ T cells were shown in a dot plot with CFSE on the x-axis, and the percentage of proliferated CD4+ and CD8α+ T cells were measured. Fig. 1B shows the results from one representative sample.

All independent predictor candidates were transformed into variab

All independent predictor candidates were transformed into variable-dependent tertile numbers, which were arranged in such a manner that a high tertile number was considered unfavourable

in terms of CD4 loss. In univariate analysis, the E/G and E/G neg ratios were not only the strongest predictors of current CD4 change rate, but in this limited cohort also the only significant predictors. For example, the odds ratio for rapid CD4 loss was 8·0 between patients within the lowest and the highest tertile of E/G ratios (i.e. 4·0 × 2 Palbociclib cost tertiles, Table 4). CD38 expression and Gag-specific CD8+ responses per se were also predictive for high relative and guideline-restricted CD4 loss rates, in contrast to HIV-RNA and β2-microglobulin (Table 4). No significant results in multivariate binary regression model were found. Clinical evaluation of asymptomatic and untreated HIV-infected patients should be based upon prognostic markers with sufficient statistical power for individual counselling. HIV-RNA levels, for selleck compound example, correlates clearly with clinical progression in large cohorts but predicts progression poorly at

the individual level [11–13]. Thus, optimal markers of progression should provide significant information even in small cohorts. This explorative study investigated new parameters for HIV-specific immunity in the search for optimal prognostic markers. The main goals of this study were to investigate prognostic significance of HIV-specific T cell responses to Gag, Env and Nef and of PD-1 on such HIV-specific cells. Specific clones were detected through transient expression of CD107a and CD154. These data were compared to quantitative measurements of CD38 on CD8+ and CD8+CD38+PD-1+ Thiamet G T cells and correlated subsequently to progression, which in asymptomatic patients may be best described by CD4+ T cell loss rates. Furthermore, fresh blood samples as opposed to thawed PBMC were analysed due to the decay of CD38 on thawed PBMC [14], possible preferential loss of CD8+ T cells [38] and limited robustness of the CD107a assay.

Two mainly affirmative observations were made: a predominance of Gag-restricted CD8+ T cell responses and their relation to prognosis [20] and a high expression of PD-1 molecules on such HIV-specific CD8+ T cells [30]. In addition, this study provided new data showing up-regulated PD-1 on HIV-specific CD4+ T cells, but differently than on the CD8+ subset as well as a lower expression of PD-1 on Env-specific CD8+ T cells compared with Gag-specific cells (Fig. 1a). Subsequently, the data on relative and absolute abundance of HIV-specific responses, including the estimates of PD-1, were related to CD4 loss rates. The total number of Gag-specific CD8+ cells were correlated even stronger with CD4 loss rates and immune activation than the conventional frequency estimates (Table 3) supporting the relevance of taking the CD8+ T cell count into consideration.

IL-5 and GM-CSF were determined in supernatants using specific EL

IL-5 and GM-CSF were determined in supernatants using specific ELISA

Kit assays (eBiosciences). The results are expressed as the mean±SD. Data were analyzed using Student’s t-test (Prism SCH772984 mw GraphPad Software, San Diego, CA, USA). This work was supported by grants from the Italian Ministry of Health, Associazione Italiana Ricerca sul Cancro, Ministero dell’Istruzione, Università e Ricerca (PRIN 2005), Fondazione Cariplo, Agenzia Spaziale Italiana (Progetto OSMA), LR.26 del Friuli Venezia Giulia. The authors thank Silvia Piconese and Mario Colombo (Istituto Tumori, Milan, Italy) for providing OX40-deficient Tregs. They are grateful to Francesco Vitrani for helpful suggestions. Conflict of interest: The authors declare no financial or commercial conflict of interest. Detailed facts of importance RXDX-106 cost 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. “
“Interleukin-19 (IL-19) plays an important role in asthma by stimulating T helper type 2 (Th2) cytokine production. Interestingly, IL-4, a key Th2 cytokine, in turn up-regulates IL-19 expression in bronchial epithelial cells, so forming a positive feedback loop. In atopic dermatitis (AD), another Th2 disease closely related to asthma, IL-19 is up-regulated in the skin. We propose to use IL-4 transgenic (Tg) mice and human keratinocyte culture to delineate the molecular mechanisms involved in the up-regulation

of IL-19 in AD. IL-19 is similarly up-regulated in the skin of IL-4 Tg mice as in human AD. Thiamet G Next we show that IL-4 up-regulates IL-19 expression in keratinocytes. Interestingly, the up-regulation was suppressed by a pan-Janus kinase (Jak) inhibitor, suggesting that the Jak–signal transducer and activator of transcription (Jak-STAT) pathway may be involved. Dominant negative studies further indicate that STAT6, but not other STATs, mediates the up-regulation. Serial 5′ deletion of the IL-19 promoter and mutagenesis studies demonstrate that IL-4 up-regulation of IL-19 in keratinocytes involves two imperfect STAT6 response elements. Finally, chromatin immunoprecipitation assay studies indicate that IL-4 increases the binding of STAT6 to its response elements in the IL-19 promoter. Taken together, we delineate the detailed molecular pathway for IL-4 up-regulation of IL-19 in keratinocytes, which may play an important role in AD pathogenesis. “
“The in vivo or in vitro formation of IgG4 hybrid molecules, wherein the immunoglobulins have exchanged half molecules, has previously been reported under experimental conditions. Here we estimate the incidence of polyclonal IgG4 hybrids in normal human serum and comment on the existence of IgG4 molecules with different immunoglobulin light chains.

We assayed bacterial burdens in the liver and kidney (Fig 4J and

We assayed bacterial burdens in the liver and kidney (Fig. 4J and K). Cav1 KO mice showed significantly increased CFUs in the liver (p = 0.001) and kidney (p < 0.001) as compared with WT mice. This result indicates that more severe dissemination occurred in cav1 KO mice than in WT mice. We studied the regulatory mechanism underlying the susceptibility

to K. pneumoniae infection in cav1 KO mice. Using western Selleckchem CDK inhibitor blotting, we found that the GSK3β−β-catenin−Akt pathway may be involved in controlling K. pneumoniae infection. The protein levels of GSK3β and IL-12a, as well as phosphorylation of Akt, GSK3β, and ERK1/2, were significantly elevated in cav1 KO mice following K. pneumoniae infection, while the protein levels of Akt, β-catenin, and STAT5 (also p-STAT5) were markedly downregulated (Fig. 5A and B, and densitometry analysis, Fig. 5C). Thus, the decreased levels of STAT5 and Akt, as well as increased levels of IL-6 and IL-12a, may result from the loss of Cav1′s negative feedback mechanism. These data suggest that the STAT5 pathway may be downregulated by a negative signal from the GSK3β − β-catenin − Akt axis in this model. Since the early time point showed altered cytokine responses, we next selleck chemicals evaluated relevant cell signaling proteins at 8-h postinfection. Our data (Fig. 5D and E) demonstrate that the cell signaling pattern at

8 h postinfection is also altered in cav1 KO mice versus WT mice by infection. Importantly, the major

responsive proteins (e.g. Akt, β-catenin, KC, and STAT5) at 8 h showed similar decreases, while other signaling proteins (GSK3β and IL-12a) did not display the increases seen at 24 h. These data were densitometrically analyzed as shown in Fig. 5F. Thus, the cell signaling data at early time points are in-line with the signaling results at late time points. However, as not all increases/decreases were the same at 8 and 24 h, our data also indicate that the cytokine responses may increase as the disease progresses. The expression of Akt and STAT5 was also measured in lung tissue using immunohistochemistry, which showed decreased staining for both proteins in cav1 KO mice versus WT mice after infection Unoprostone (Fig. 5G, arrows indicating significant changes in fluorescent intensity between control and KO mice lungs). As previous studies show that GSK3β can destabilize β-catenin [[17]], we speculate that GSK3β may negatively regulate Akt or β-catenin, leading to a lowered STAT5 and dysregulated cytokine patterns. Since IL-27 has previously been shown to be associated with STAT1, we also evaluated the expression levels of STAT1, and found that there were no significant differences between control mice and KO mice (data not shown). Similar changes in β-catenin, GSK3β, and cytokine (IL-6 and IL-12a) levels were observed in lung tissue of cav1 KO mice as assessed by immunostaining (Supporting Information Fig. 1 and 2).

The rank order of OAB prevalence rate of patients with each backg

The rank order of OAB prevalence rate of patients with each background disease was 40.0% (ischemic heart disease), 36.5% (brain and neurological disease), 34.9% (psychiatric disease), 32.8% (gastrointestinal disease), 32.1% (diabetes mellitus), 27.4% (hypertension), 25.7% (hyperlipidemia), 24.3% (orthopedic disease),

GW-572016 cost 18.5% (respiratory disease) and 17.0% (gynecological disease). To evaluate of the contribution of each disease to the OAB prevalence rate, multiple regression analysis was performed. The analysis showed that ischemic heart disease, brain and neurological disease, psychiatric disease, hypertension, gastrointestinal disease and diabetes mellitus have significantly higher odds ratios for the OAB prevalence rate (Table 1). There is evidence showing close association between lower urinary tract symptoms (LUTS) and major chronic medical diseases as well as related lifestyle factors.8 Furthermore, higher concentration of oxidized LDL was associated with increased incidence of metabolic syndrome overall, as well as its components of abdominal obesity, hyperglycemia, and hyperlipidemia.9 These data suggest that it might be possible that hyperlipidemia is one of important factors for LUTS,

including OAB. However, in this study, hyperlipidemia did not show the significant contribution Acalabrutinib clinical trial to OAB prevalence rates. Further studies will be needed to clarify this reason. WHHL rabbits were first reported in 1980 as a strain of rabbit with a constantly inherited hyperlipidemic trait produced by inbreeding from a mutant

discovered in 1973,10 and later their hyperlipidemia was found to be due to reduced LDL function derived from an in-frame deletion of 12 nucleotides that eliminates four amino acids from the cysteine-rich ligand binding domains of the LDL receptor.11 Since 1994, the development of an animal model for spontaneous myocardial infarction by serial and selective breeding of the coronary atherosclerosis–prone WHHL rabbits has been attempted. After 6 years of selective breeding, a new WHHL strain for spontaneous myocardial infarction was developed, and was named myocardial infarction-prone WHHL rabbit strain ADP ribosylation factor (WHHL-MI rabbit). In WHHL-MI rabbits, there is a higher fraction of low-density lipoprotein (LDL) in hyperlipidemic rabbits than in the control rabbits. High level of LDL cholesterol is one of the risk factors for arterial infarction. In addition, it has been reported that higher level of oxidized LDL cholesterol contributes to higher incidence rate of metabolic syndrome.11 Aortic atherosclerosis in WHHL-MI rabbits is observed grossly from 2 months of age, despite being fed normal chow, and at 12 months of age, atherosclerosis covers about 70% of the aortic surface.

Excess p53-binding nucleotide, which does not contain a GAS seque

Excess p53-binding nucleotide, which does not contain a GAS sequence, did not compete-out the binding of STAT1. check details Therefore, our data suggest that constitutive STAT1 binding to the GILT promoter occurs at GAS sites. In addition, we tested whether mutations that affect the activity of the GILT promoter can influence in vitro binding to the GAS sites in the GILT promoter. The results shown

in Fig. 2b indicate that mutant K544A/E545A (Mut 3) binds to the GILT promoter but mutant V426D/T427D (Mut 1) does not bind GAS sequences in GILT promoter, as expected. However, repeated DAPA did not detect binding of E428A/E429 (Mut 2), although this mutant behaved like STAT1α in the luciferase assay. This may be a result of either the limit of detection of DAPA or because this mutant exerts its

effect on the GILT promoter indirectly. To determine whether mutant STAT1 interacts with the specific sequences in the GILT promoter, regardless of the phosphorylation, WT, Stat1−/−, Stat1β-Y701 and Stat1α-S727 MEFs were treated with IFN-γ and the lysates were incubated with biotinylated PLX-4720 order oligonucleotides of Stat1 Probe 1 and Probe 2 (Fig. 4a). Our data indicate that, regardless of phosphorylation of Y701 and S727, STAT1 is able to bind target sequences in the GILT promoter. However, to confirm that what is seen here is specific binding, lysates from Stat1−/− cells transfected Oxaprozin transiently with Stat1α, Stat1β-Y701 and Stat1α-S727 were incubated with biotinylated oligonucleotides

of Stat1 Probe 1 and Probe 2 (Fig. 4b). The reactions were competed-out with a 50-fold excess of unlabelled probe corresponding to either Stat1 consensus or p53 sequences. Our data indicate that WT and Stat1 mutants can bind specifically to the sequences in the GILT promoter. Similar results were achieved with the Stat1 probe 1 (data not shown). During an early immune response the expression of various immune molecules is induced. GILT is constitutively expressed in professional APCs and is also inducible in vitro in APCs by inflammatory cytokines such as IFN-γ, tumour necrosis factor-α (TNF-α) and interleukin-1β (IL-1β). Stat1 has been shown to regulate the IFN-γ-stimulated induction of GILT.12 However, we found that GILT is also constitutively expressed at detectable levels in other cell types not involved in antigen processing, such as mouse T cells and skin fibroblasts.9,10 Therefore, GILT is produced at basal levels without any extracellular stimuli. We were interested to determine whether Stat1 plays any role in the constitutive expression of GILT. We expected that the absence of Stat1 in Stat1−/− cells would reduce the expression of GILT. Surprisingly, the Stat1−/− mouse fibroblast cell line (MEF) showed increased levels of GILT protein, suggesting that STAT1 may exert a negative regulation on the constitutive expression of GILT.

Undoubtedly, the most studied factor in Echinococcus is the so-ca

Undoubtedly, the most studied factor in Echinococcus is the so-called antigen B (AgB), a highly immunogenic lipoprotein and major component of hydatid cyst fluid (94). Although

there are several reports on Silmitasertib in vivo immunomodulatory properties of AgB in vitro (94), and biochemical investigations that demonstrate binding of different hydrophobic ligands to AgB (95), the precise function of this protein in the biology of Echinococcus or in the immune response during echinococcosis is still unknown. Originally described as a 160 kDa lipoprotein, AgB was later shown to be built up of several 8 kDa monomers that are encoded by a gene family (96), and since the first full description of an AgB-encoding gene by Frosch et al. (97), there has been constant debate on how many of these genes are actually A-769662 research buy expressed in these parasites. By studies of Fernandez et al. (98), Chemale et al. (99), Arend et al. (100) and Mamuti et al. (101), the number of AgB subunit genes had grown to five in 2007 (named EmAgB1-EmAgB5 in E. multilocularis and EgAgB1-EgAgB5 in E. granulosus), whereas genomic Southern blot analyses indicated that there are at least seven loci

(102). Studies by Haag et al. (103) and Arend et al. (100) even suggested the presence of further AgB genes (up to 10 in E. granulosus and up to 110 copies in the related E. ortleppi) as well as a high degree of genetic polymorphism among those genes (even within protoscoleces that derived from one single cyst). These authors proposed that numerous AgB copies might be involved in gene conversion mechanisms through recombination processes and DNA rearrangements similar to the situation in protozoans such as Plasmodium sp. or trypanosomes (103). This theory was recently contradicted by Zhang et al. (104) who characterized AgB genes in E. granulosus isolates from different geographic origins and proposed the presence of 10 unique genes (or alleles) that are, however, highly homologous between these isolates and did not

show gross polymorphisms. To shed more light on the situation, we have Bupivacaine analysed the presence and location of AgB genes in the current assemblies of the E. multilocularis and E. granulosus genomes. As described by Brehm (72), using the first assembly version of the E. multilocularis genome (19 000 contigs), a total of seven AgB loci appears to form a cluster on a distinct region of the genome. In the latest genome version (600 supercontigs), all these copies are now assembled into one continuous sequence fragment of 57 kbp that is present on scaffold_29 (Figures 2 and 3). The antigen B cluster is flanked by two genes, EmLDLR and EmMTA, which are highly conserved among cestodes.

Despite the lack of TFH cells and GCs in these mice, memory B cel

Despite the lack of TFH cells and GCs in these mice, memory B cells still developed, consistent with a GC-independent pathway. However, it also suggested that this pathway is independent of TFH cells. T cell help and CD40/CD40L interactions are required for both GC-dependent and GC-independent memory B cell formation, as in the absence of the costimulatory molecule CD40L neither developed. In conclusion, this shows that the early GC-independent and late GC-dependent memory B Rapamycin cells develop aided by different T helper cell subsets. Ti B cell responses can be

divided into two main groups Ti-1 and Ti-2 based on the type of antigen. Ti-1 antigens, for example, bacterial lipopolysaccharide (LPS), possess an intrinsic activity that can directly induce B cell activation regardless of antigen specificity, and they also provide selleck chemical the B cell with a second signal via Toll-like

receptors. Ti-2 antigens, for example, pneumococcal polysaccharide or the model antigen 2,4-dinitrophenyl coupled to dextran (DNP-DE), are highly repetitive structures that cross-link a sufficient number of BCRs to fully activate antigen-specific B cells. Ti-1 antigens can activate both immature and mature B cells, while Ti-2 antigens only activate mature B cells. Ti-2 B cell responses are mainly executed by B1 and MZ B cells [40] and are localized to extrafollicular triclocarban foci [41]. For many years, it was believed that responses against Ti antigens could not give rise to immunological memory. Early studies showed that rechallenge with DNP-DE after primary immunization induced a poor anti-DNP antibody response. However, this unresponsiveness was not due to a lack of antigen-specific memory B cells but rather to the production of hapten-specific antibodies that inhibited B cell triggering [42, 43]. In support of this, adoptive transfer of DNP-DE-primed spleen cells to irradiated recipients followed by rechallenge, resulted in an enhanced IgM

response [44]. More recently, it has been shown that B1b cells give rise to memory B cells in response to Ti antigens [45], and also, B1a cells appear to develop memory-like features [46, 47]. Ti memory B cells appear phenotypically different with respect to certain markers compared with Td B memory cells [43]. Autoantibodies are present in mouse models of autoimmune diseases such as systemic lupus erythematous (SLE), type I diabetes and rheumatoid arthritis (RA) and contribute to the pathogenicity. However, production of autoantibodies per se does not necessarily induce autoimmune disease [48], rather the complex pathological manifestations of these diseases are under the control of combinations of multiple genes [49].

Concentrations of IL-4 (R&D Systems, Minneapolis, MN, USA), IL-10

Concentrations of IL-4 (R&D Systems, Minneapolis, MN, USA), IL-10 (Pierce Biotech Inc., Rockford, IL, USA), IL-12 (Pierce Biotech Inc.) and IL-13 (R&D Systems) in the supernatants were quantified using commercial ELISA kits according to the manufacturer’s instructions. Quantitation of mRNAs of PARs by real-time PCR. 

Expression of PAR mRNAs in P815 cells was determined by real-time PCR FK506 as described previously [8]. Briefly, real-time PCR was performed by using SYBR®Premix Ex TaqTM on the ABI Prism 7700 Sequence Detection System (Perkin Elmer Applied Systems, Foster City, CA, USA). The sequences of the primers are summarized in Table 1. PAR-1, PAR-2, PAR-3 and PAR-4 mRNA expression in each sample was finally determined after correction with β-actin expression. Flow cytometry and immunofluorescent microscopy analyses of PARs.  The staining procedures were mainly adopted from the one described previously for Per a 7 [8]. Cells were then analysed on buy PCI-32765 a FACS Calibur flow cytometer with CellQuest software (BD Biosciences, San Jose, CA, USA) or on a Nikon EZ-C1 confocal laser-scanning microscope (Japan). Statistical analysis.  Data were expressed as mean ± SEM for four independent experiments. Where analysis of variance indicated significant differences

between groups with ANOVA, for the preplanned comparisons of interest, Student’s t test was applied utilizing the spss 13.0 version (SPSS Inc., Chicago, IL, USA). P < 0.05 was taken as statistically

significant difference. In order to investigate the functions of Per a 1.01, we prepared rPer a 1.0101 and rPer a 1.0104. The E. coli generated approximately 82 and 23 mg/l Epothilone B (EPO906, Patupilone) culture mixture rPer a 1.0101 and rPer a 1.0104 proteins respectively, which consisted of approximately 24% of total soluble bacterial proteins (Fig. 1A). After purification, the recombinant proteins with apparent molecular weights 28 and 33 kDa were observed on a SDS–PAGE (Fig. 1B). Solubility analysis showed that Per a 1.0101 and Per a 1.0104 possessed very high probabilities (>90%) of being soluble when expressed in E. coli. In order to ensure our recombinant proteins are Per a 1.0101 and Per a 1.0104 molecules, we examined the proteins by LC-ESI-MS/MS analysis. Following trypsin digestion, seven peptide fragments from Per a 1.0101 and 4 peptide fragments from Per a 1.0104 (Table 2) were obtained. They matched well with Per a 1.0101 and Per a 1.0104 protein sequences. As large numbers of allergens possess enzymatic activities [14, 15], we examined tryptic, chymotryptic, metalloproteinase and aspartic proteinase activities of purified rPer a 1.0101 and rPer a 1.0104. At the concentrations of 0.5, 5.0 and 50 μg/ml, rPer a 1.0101 and rPer a 1.0104 failed to show any tryptic or chymotryptic metalloproteinase and aspartic proteinase activities towards substrates BAPNA, SAAPP, casein and haemoglobin, respectively. To confirm Per a 1.0101 and Per a 1.

The two groups of recipient mice produced low levels of antibody

The two groups of recipient mice produced low levels of antibody in serum 4 weeks after transfer of BMDC and no significant difference in antibody response was observed between the two groups (Fig. 7a). However, OVA antigen boosting 4 weeks after BMDC transfer enhanced the antibody responses. Mice receiving BMDC that were treated with rHp-CPI and pulsed with OVA produced significantly less OVA-specific total learn more immunoglobulin and IgG1 than the mice that received BMDC pulsed with OVA antigen only (Fig. 7b). No significant levels of IgG2a antibody were detected in the BMDC recipient

mice and the mice injected with OVA antigen only (Fig. 7b). These data show that rHp-CPI is able to modify the DC phenotype and function resulting in impaired antibody response. Immunosuppression that occurs following infection with murine nematode H. polygyrus has been documented extensively.[33-35] The H. polygyrus-derived ES products have been shown to induce immunosuppression in hosts by impairing DC function.[15] However, the parasite molecule(s) responsible for induction of immunosuppression are unknown. In this

study, we cloned the CPI gene from H. polygyrus, produced recombinant protein rHp-CPI and examined its immunomodulatory effects. Our results demonstrated that the SB203580 concentration recombinant rHp-CPI protein is biologically functional as shown by its ability to inhibit the protease activity of a panel of cathepsins. Immunoblotting assays revealed that the mAb raised against the rHp-CPI protein was able to recognize a protein component in H. polygyrus ES products, indicating that H. polygyrus produces Phosphatidylinositol diacylglycerol-lyase and secretes the CPI protein. Indeed, the ES products prepared from H. polygyrus adult worms showed inhibitory activity against cathepsins (Fig. 2). There are several reports to show that

nematode parasites that dwell in the gastrointestinal tract of their hosts are able to modulate the immune response systemically.[21, 36] In a previous study, we have shown that concurrent H. polygyrus infection impairs protective immunity against systemic malarial infection.[24] A study by Goodridge et al.[32] showed that the immunomodulatory glycoprotein ES-62 of a filarial nematode released by an osmotic pump implanted in the neck of mice is able to induce hyporesponsive DC derived ex vivo from the bone marrow cells of mice. These observations suggest that the immunomodulatory molecules released by adult H. polygyrus may modulate the functions of immune cells locally as well as in other organs of the immune system, including bone marrow where the DC progenitors differentiate and develop into immature DC. To verify this possible mechanism, bone marrow cells were cultured in the presence of rHp-CPI and the phenotypes of the differentiated CD11c+ DC were analysed.