In vitro treatment of B-1 B cells with hapten–protein

In vitro treatment of B-1 B cells with hapten–protein Selleckchem Gemcitabine complex.  Naïve wild-type B-1 B cell-containing

peritoneal cells were incubated with the hapten–protein complex trinitrophenyl–bovine serum albumin (TNP–BSA, prepared at a concentration of 50 μg/ml in RPMI 1640 culture media supplemented with 5% FCS) for 40 min at 37 °C. Incubation of iNKT cells with B-1 B cells.  B-1 B cell-containing peritoneal cells exposed to TNP-BSA and iNKT cell-containing LMNC exposed to lipid extracts were washed and co-incubated in vitro for 40 min at 37 °C. Centrifuged pellets of the activated iNKT and B-1 B cell mixture were resuspended in PBS prior to adoptive transfer. Adoptive transfer.  To reconstitute iNKT cells in Jα18−/− or CD1d−/− mice, we transferred LMNC into Jα18−/− or CD1d−/− mice at a dose of 0.5–1 × 106 cells per mouse. To reconstitute B-1 B cells, we transferred the mixture of peritoneal cells and LMNC into JH−/− or CBA/N-xid mice at a dose of 5 × 106 cells per mouse. Cells were transferred via intravenous injection into the retro-orbital plexus of recipient mice under methoxyflurane anaesthesia 1 day prior to challenge (i.e., day 3 after sensitization). Flow cytometry with CD1d-α-GalCer Selleck JNK inhibitor tetramers.  Liver mononuclear cells were washed and resuspended in PBS staining buffer containing 2% BSA, stained with a mixture of FITC-anti-TCR-β antibody and PE-labelled CD1d-α-GalCer

tetramers on ice for 30 min and washed twice more. The double-positive cells (iNKT cells) were identified using a FACS Calibur flow cytometer (Becton Dickinson, Franklin Lakes, NJ, USA) and reported as a percentage of total αβ-TCR-positive LMNC (T cells). iNKT cells constitute

approximately 70% of hepatic T cells in the wild-type H-2d mice employed here. Results were analysed using Mac CellQuest (BD). Isolation and flow cytometry of for hepatocytes.  Mice were anesthetized with intra-peritoneal pentobarbital before entering the abdomen. Portal veins were perfused with Hanks A solution for 3–4 min and Hanks B solution with collagenase until signs of liver digestion became apparent. The livers then were removed. The hepatocyte fraction was strained through a 70-m mesh (BD) and stained with FITC-anti-CD1d antibody for 1 h on ice before analysis by flow cytometry. Results were analysed using Mac CellQuest (BD). It is not understood how iNKT cells respond so rapidly to contact sensitization. Our hypothesis was that the character of hepatic lipids changes in a manner that increases their capacity to stimulate iNKT cells. To investigate this, we utilized adoptive transfer techniques in JH−/− and CBA/N-xid mice, which lack B cells and B-1 B cells, respectively. Both strains thus have impaired CS at baseline at both 2 and 24 h after challenge (Group B in Fig. 1A,B). We previously demonstrated that CS is impaired in these B cell–deficient mice compared with wild-type mice and that CS could be fully reconstituted with adoptive transfer of sorted B-1 B cells previously activated in vivo [8, 10].

Total

Total BAY 80-6946 RNA was extracted from cells or tissues using Isogen (Nippon

Gene, Tokyo, Japan). Single-strand cDNA was synthesized using ExScript RT reagent kits (Takara, Otsu, Japan). Real-time RT–PCR was performed using an ABI PRISM 7500 Sequence Detection System (Applied Biosystems, Foster City, CA, USA), with primers described in Table 1. Amplifications were performed in duplicate with SYBR Premix Ex Taq (Takara), according to the manufacturer’s instructions. Target mRNA levels were normalized against β-actin mRNA. Bone marrow dendritic cells (BMDC) were obtained from WT or FcγRIIb-deficient mice according to the method described previously [18]. The bone marrow cells were cultured at 1 × 106 cells/ml in the presence of 20 ng/ml BAY 73-4506 murine granulocyte–macrophage colony-stimulating factor (GM-CSF). The medium was replaced with a GM-CSF-containing medium on day 4 of culture. On day 6 of culture, BMDCs were collected and CD11c+ BMDCs were purified using the autoMACS system. Sensitized FcγRIIb-deficient mice were injected i.v. with 1 × 106 CD11c+ BMDCs 24 h before i.v. administration of IgG and challenged with OVA for 3 days. All results are expressed as mean ± standard deviation. A t-test was conducted

to determine differences between two groups. As measured values were not distributed normally and the sample size was small, non-parametric analysis using a Mann–Whitney U-test confirmed that differences remained significant, even if the

underlying distribution was uncertain. The P-values for significance were set at 0·05 for all tests. To estimate the effects of IVIgG on bronchial asthma, rabbit IgG was administered intravenously to the murine allergic airway inflammation model. OVA sensitization and challenge induced a substantial increase Selleck Fluorouracil in total cells in BALF. This was due largely to increased eosinophil numbers, which is one of the characteristics of eosinophilic airway inflammation in bronchial asthma. Administration of 1 mg of rabbit IgG before airway challenge markedly decreased the number of total cells and eosinophils in BALF (Fig. 1a) in a dose-dependent manner. The treatment, such as the same amount of IgM or F(ab′)2, did not influence significantly the BALF cell counts, nor did administration of 1 mg of mouse IgG influence cell counts. In the IVIgG experiment after challenge, rabbit IgG administration after OVA challenge for 3 days also reduced the number of total cells and eosinophils significantly compared with PBS-treated mouse (Fig. 1b). Because 1 mg of rabbit IgG suppressed airway inflammation sufficiently, we used this dose to analyse the role of IVIgG before OVA challenge in our subsequent experiments. Plasma OVA-IgE levels were also elevated in challenged mice. This effect was suppressed by rabbit IgG administration (Fig. 1c). Next, to assess the effect of IVIgG on AHR, the relative increase of Penh in response to methacholine inhalation was evaluated.

g reactive oxygen species) and non-oxidative (e g various prote

g. reactive oxygen species) and non-oxidative (e.g. various proteases) mechanisms.[17] The importance of neutrophil function is evident in individuals who have defects in neutrophil chemotaxis,

phagocytic functions or who have neutropenia.[18, 19] These individuals are more prone to bacterial infections. On the other hand, microbicidal molecules released from activated and dying neutrophils can cause bystander damage Sirolimus in vitro to healthy tissue. The consequent cell injury and death can itself cause or aggravate disease. Accordingly, it is important to elucidate the factors controlling neutrophilic inflammation. In this study we describe the surprising finding that the gut flora influences the ability of animals to mount a systemic acute neutrophilic inflammatory response

in the peritoneum and characterize the underlying basis for this observation. Specific pathogen free (SPF) C57BL/6 mice and IL-1R−/− mice were purchased from The Jackson Laboratories (Bar Harbor, ME). Germ-free C57BL/6 BMS-907351 molecular weight mice were obtained from The National Gnotobiotic Rodent Resource Center, North Carolina State University Gnotobiotics Unit and Gnotobiotic Research Resource, Medical University of South Carolina. MyD88−/− mice were provided by Dr Shizuo Akira, Osaka University, Osaka, Japan or purchased from The Jackson Laboratories. RIP2−/− mice were provided by Dr Michelle Kelliher and RIG-I−/− and MDA5−/− mice were provided by Dr Kate Fitzgerald (University of Massachusetts Medical School, Worcester, MA). NOD1−/− mice were Chlormezanone provided by Dr Grace Chen, University of Michigan, Ann Arbor, MI. For generating the tamoxifen-inducible deletion mutant mice of MyD88, we used a strategy similar to the one described

previously.[20] MyD88−/− mice were crossed to the whole tissue, tamoxifen-inducible Cre transgenic mice (Rosa26-Cre/ESR+/+) (provided by Dr Roger Davis, University of Massachusetts Medical School, Worcester, MA). The resultant offspring, MyD88+/− Rosa26-Cre/ESR+/− mice were crossed to the MyD88flox/flox mice (provided by Dr Robert Finberg, University of Massachusetts Medical School, Worcester, MA) to generate the MyD88−/flox Rosa26-Cre/ESR+/− (conditional knockout; cKO). Animals were housed and handled according to protocols approved by the University of Massachusetts animal care and use committee. Mice were injected intraperitoneally with 0·2 mg zymosan (Sigma-Aldrich, St Louis, MO), 0·5 mg silica crystals (Sigma-Aldrich), 0·5 mg monosodium urate crystals or 5 ng recombinant murine MIP-2 (R&D Systems, Minneapolis, MN) in 0·2 ml PBS. For the thioglycollate injections, 1 ml of 3% thioglycollate (Thermoscientific, Lenexa, KS) was used. The monosodium urate crystals were prepared as described before.[21] Mice were killed by exposure to isoflourane 4–16 hr after the injection. The peritoneum was lavaged with 2 ml Dulbecco’s modified Eagle’s medium with 2% fetal calf serum, 3 mm EDTA and 10 U/ml heparin.

All animal experiments described in the paper were done under UK

All animal experiments described in the paper were done under UK Home Office Project Licence numbers 70/5791 and 70/6724 and were approved by the in-house ethics committee of the Institute of Cancer Research. All antibodies for flow cytometry were purchased from eBioscience or BD Biosciences. The following fluorescently labeled or biotin-conjugated anti-mouse antibodies were used: CD4 (GK1.5), CD69 (H1.2F3), CD8α(53-6.7), CD8β (CT-CD8b), TCR-β VX-765 (H57-597), CD5 (53-7.3), Bcl-2 (3F11), IL-7Rα (B12-1). Staining by biotin-conjugated antibodies was visualised using streptavidin-conjugated

fluorophores. Immunofluorescence data were collected using a Becton Dickinson FACSCalibur, or a Becton Dickinson LSRII using CellQuest software and analysed LY2157299 mouse using Flowjo software (Treestar). Cells were sorted on a FACS Aria (Becton Dickinson) or a MoFlo (DakoCytomation), with DAPI staining used to exclude dead cells. Total RNA was

isolated using Trizol Reagent (Invitrogen) according to the manufacturer’s instructions. cDNA was synthesised using Invitrogen M-MLV Reverse Transcriptase. Each reaction contained 200 U enzyme, in 50 mM Tris-HCl (pH 8.3), 75 mM KCl, 3 mM MgCl2 and 10 mM DTT, 1 mM dNTP, 500 ng oligo (dT)15 primer (Promega), and 40 U RNAsin ribonuclease inhibitor (Promega) and was incubated for 50 min at 37°C, prior to heat inactivation at 70°C for 15 min. For qPCR, gene-specific primer/probe sets were purchased from Applied Biosystems as “Gene Expression Assays”, and reactions were performed with Taqman Universal PCR Mastermix (Roche/Applied Biosystems) on an ABI 7900 Real Time PCR System, using Hprt or Rps16 as a comparator. Standard curves were created using standards (usually serial dilutions of total thymus cDNA) for relative quantitation of the data. To assay the kinetics of Egr2 upregulation, MHC° thymocytes were cultured with 10 ng/mL PMA and 1 μg/mL Ionomycin. Signaling inhibitors were included at Lenvatinib cell line the following concentrations: U0126 (Promega) 10 μM, PD98059 (Promega)

10 μM, cyclosporin A (Calbiochem) 50 nM, FK506 (Sigma) 1 nM. For Erk phosphorylation in response to TCR ligation, thymocytes were treated with anti-CD3 diluted 1/100 in PBS, then warmed to 37°C. Goat anti-Armenian Hamster IgG (75 μg/mL; Jackson ImmunoResearch) was added and the cells were left for 2 min at 37°C before addition of paraformaldehyde to a final concentration of 2%, and incubation on ice for 10 min. Following centrifugation, cells were resuspended in 90% methanol and incubated for 30 min. Permeabilised cells were stained with Phospho-p44/42 MAPK (Thr202/Tyr204) (E10) Mouse mAb (Alexa Fluor 488 Conjugate) from Cell signaling Technology, using PBS-0.5% BSA as the staining buffer, in accordance with the manufacturer’s instructions. For anti-CD3 stimulation, 48-well tissue culture plates were coated with 150 μL of 2 μg/mL anti-mouse CD3ε (145-2C11) in PBS and incubated overnight at 4°C.

The Fli-1+/− MRL/lpr mice receiving BM from WT MRL/lpr mice also

The Fli-1+/− MRL/lpr mice receiving BM from WT MRL/lpr mice also had improved disease development compared to WT MRL/lpr mice that received BM from WT MRL/lpr mice. These findings indicate that the impact of Fli-1 on disease development in MRL/lpr mice is complex, and involves both haematopoietic cell and non-haematopoietic cell mediated mechanisms Fli-1+/− MRL/lpr mice were generated as described previously [13]. WT MRL/lpr mice were purchased from the Jackson

Laboratory (Bar Harbor, ME, USA). Fli-1+/− MRL/lpr mice used in this study were back-crossed with WT MRL/lpr mice for 12 generations. The major histocompatibility complex (MHC) locus for MRL/lpr Fli-1+/− mice was the same this website as in WT MRL/lpr mice. Two groups of mice, WT MRL/lpr and Fli-1+/− MRL/lpr, were generated by breeding Fli-1+/− MRL/lpr mice with WT MRL/lpr mice. Mice were examined twice-weekly for external disease manifestations such as skin rash, ear necrosis and lymph node enlargement. All mice were housed under pathogen-free MK-8669 datasheet conditions at the animal facility of the Ralph H. Johnson Veterans Affairs Medical Center. Four groups of 10-week-old MRL/lpr mice (10–12 mice/group) were irradiated with fractionated irradiation (5 Gy X2; 4-h interval). Three

h after final irradiation each mouse in the four groups received 1 million BM cells by tail vein injection. In group 1, WT MRL/lpr mice received BM from Fli-1+/− MRL/lpr mice (Fli-1+/− WT). In group 2, Fli-1+/− MRL/lpr mice

received BM from WT MRL/lpr mice (WT Fli-1+/−). In group 3, WT MRL/lpr mice received BM from WT MRL/lpr mice (WT WT). In group 4, Fli-1+/− MRL/lpr mice received BM from Fli-1+/− MRL/lpr mice (Fli-1+/− Fli-1+/−). BM cells collected from donor mice at the age of 8 weeks. To monitor the efficiency of irradiation, eight WT MRL/lpr mice were irradiated as above without receiving BM transplantation. This total body irradiation was performed using a 6 × 106 eV linear accelerator (Clinac 600, Varian, Palo Alto, CA, USA). BM cells were flushed from femurs using Alpha modified Eagle’s medium (MEM) without deoxyribosides and ribosides, supplemented with 0·1% bovine serum albumin (BSA), penicillin and streptomycin (MP Biomedicals, Aurora, OH, USA). The sex of BM cell donors was mismatched second to receivers to determine the efficiency of BM transplantation. All irradiated mice were treated with 1 mg/ml neomycin sulphate for 3 weeks while in recovery from the BM transplantation. Sera were collected from the four groups of mice 12 weeks after BM transplantation at 4-week intervals. Mice were killed at 24 weeks after BM transplantation for assessment of renal disease. BM transplantation was performed in another four groups of mice (10–12 mice/group, equal female and male) as described above, and these mice were used to assess the impact of different BM transplantation on survival.

2), indicating that cell identity was altered due to the action o

2), indicating that cell identity was altered due to the action of GM-CSF. In other words, GM-CSF changed the DC progeny of Flt3L cultured

BM progenitors. It is well accepted that GM-CSF and Flt3L mobilize different ACP-196 precursor cells for DC differentiation. Ly6Chigh monocytes are the final precursor stage en route to the generation of GM-DCs from total BM [24]; but not FL-DCs, whose immediate precursors are Ly6C− pro-DCs [22]. Therefore, the dominant nature of GM-CSF over Flt3L in BM culture raises further issues. What is the developmental fate of precursor cells for FL-DCs in the dual cytokine culture? Do FL-DCs die of neglect or is their differentiation hijacked by GM-CSF to divert fate to GM-DCs? Data both already published and uncovered in the current study support the latter scenario. First, as the Flt3L level present in the dual cytokine cultures was the same as it was in the Flt3L single cultures, there should not have been a lack of Flt3L stimulation. Second, DC progenitors, including pro-DCs, express GM-CSF receptor [16], which makes it physically possible for these cells to receive GM-CSF signaling during maturation. Third, Flt3L signaling in BM

progenitor cells activates the transcription factor STAT3, whereas GM-CSF signaling activates STAT1, STAT3, and STAT5 [25]. Therefore, when DC precursors meet both cytokines, the signaling pathway of GM-CSF can subsume that of Flt3L. Fourth, some hematopoietic cytokines, such as M-CSF and G-CSF have already been reported to govern lineage choice [26, 27]. Finally, when purified and cultured in the presence of both Flt3L and see more GM-CSF, FL-DC-committed precursor cells were diverted toward GM-DCs in spite of the presence of Flt3L. This is direct in vitro evidence for a lineage diversion role of GM-CSF. Although the incidence of macrophage colony-forming CHIR-99021 manufacturer cells remained around 5% in these pro-DCs [22], this may have

been skewed by the collection of only cells loosely attached to the substrate at the end of the culture, thus omitting most of the strongly adherent macrophage population. However, phenotypic analysis consistently indicated that the harvested cells were predominantly CD11chi and MHCII+ DCs (Fig. 5) and thus unlikely to be macrophages. Furthermore, it would be difficult to explain the more than twofold expansion in cell numbers in the cultures with dual cytokines compared with that of Flt3L alone to be due to a minor macrophage population (Fig. 5). For the above reasons, we do not think that the altered outcomes from the combined GM-CSF and Flt3L additions were due to outgrowth from distinct precursors within the enriched pro-DC population. Nevertheless, we tried to perform limiting analysis studies using GFP+ pro-DCs from mice transgenic for GFP under the promiscuous UBC promoter [15]. We seeded pro-DCs at either one or ten cells per well with 200,000 BM feeder cells in 96-well microtitre trays.

The origin seems multi-factorial, but to an important extent expl

The origin seems multi-factorial, but to an important extent explainable by prednisolone action. Gene signatures in patients with AAV undergoing steroid treatment should therefore be interpreted accordingly. “
“The endotoxic activities of lipopolysaccharides (LPS) isolated from different strains of rhizobia and rhizobacteria (Bradyrhizobium, Mesorhizobium, and Azospirillum) were compared to those of Salmonella enterica sv. Typhimurium LPS. The biological activity of all the examined preparations, measured as Limulus lysate gelation, production of tumor necrosis factor (TNF), interleukin-1β (IL-1β),

and interleukin-6 (IL-6), and nitrogen oxide (NO) induction in human myelomonocytic cells (line THP-1), was considerably lower than that of the reference enterobacterial endotoxin. Among the rhizobial lipopolysaccharides, the activities of Mesorhizobium click here huakuii and Azospirillum lipoferum LPSs were higher than those of the LPS preparations from five strains of Bradyrhizobium. The weak endotoxic activity of the examined preparations was

correlated with differences in lipid A structure compared to Salmonella. Soil bacteria belonging to the rhizobium lineage are able to fix atmospheric nitrogen during symbiosis with legume plants. Bacteria from the genus Bradyrhizobium induce nitrogen-fixing nodules on the roots of cultivated (Glycine max and Glycine soya) and wild-growing legumes (1, 2). M. huakuii induces the formation of nodules on the roots of Astragalus sinicus (3). A. lipoferum represents plant-growth-promoting rhizobacteria which colonize the root surface and are not able to penetrate root Lck cells. They live in association GPCR Compound Library purchase with roots of grasses, cereals, and other monocotyledonous plants (4, 5). Lipopolysaccharide, as an integral component of the cell walls of Gram-negative bacteria, plays an essential role in the proper development of symbiotic relationships (6). LPS, together with Omp proteins, is responsible for the asymmetric structure and semi-permeability of outer membranes. This is important for the appropriate morphogenesis and functionality of bacteroids, endosymbiotic forms of rhizobia which perform nitrogen

fixation (7). LPS may play a role in the protection of rhizobia against plant defense response mechanisms. Suppression of systemic acquired resistance or hypersensitivity reaction has been shown during infection of plant tissues by microsymbionts (8–10). Most pathogenic bacteria possess LPSs displaying endotoxic activity against host organisms. Lipid A, the part of LPSs that anchors the whole macromolecule in the outer membrane, is the centre of endotoxicity. The fine structure of enterobacterial lipid A has been identified as a glycolipid comprised of a β-(1,6)-linked glucosaminyl disaccharide substituted by two phosphate groups at positions C-1 and C-4 and six fatty acid residues with two acyloxyacyl moieties with a distinct location (Fig. 1) (11, 15, 16).

01) To further validate the in vivo findings in our aGvHD model,

01). To further validate the in vivo findings in our aGvHD model, we also tested the functional capacity of our aTreg cells to prolong allogeneic skin graft survival. As depicted in Figure 5, 1 day prior to transplantation,

C57BL/6Rag–/– mice were reconstituted with 2 × 105 CD4+CD25+ aTreg cells isolated from primary cultures together with 1 × 105 CD8+ and 1 × 105 CD4+CD45RBhigh T cells. As a control, we included a group receiving Teff cells only. aCD4+TGF-β+RA aTreg cells prolonged graft survival compared to mice reconstituted with aTreg cells from untreated or aCD4-only treated cultures (*p ≤ 0.5) (Fig. 5B). In contrast, DAPT purchase aCD4+Rapa aTreg cells did not perform better than aTreg cells obtained from aCD4-only treated cultures. Interestingly, animals receiving aCD4+TGF-β+RA aTreg cells showed also an improved recovery and weight gain after transplantation compared with mice receiving aTreg cells from all other groups (Fig. 5C). Here, we present an optimised protocol for in vitro generation of murine aTreg cells with improved in vivo function in two independent models of transplantation tolerance. This could be accomplished by addition of TGF-β+RA or Rapa Erlotinib purchase to our previously described aCD4-mAb Treg-cell expansion protocol [16]. Notably, the optimised aTreg-cell expansion

protocol increased aTreg-cell frequencies and absolute aTreg-cell counts while reducing the number of undesired Teff cells. The aTreg cells were predominantly generated by an expansion of nTreg cells. Helios and Neuropilin-1 expression levels,

stability of the aTreg phenotype, and the suppressive in vitro and in vivo function exceeded in our novel aCD4+TGF-β+RA Treg protocol over all other protocols including addition of Rapa. Several protocols for the generation alloreactive T cells with regulatory function, shown to suppress anti-donor immune responses, have been described in addition to our anti-CD4mAb-based enough protocol. These include IL-10-mediated induction of Tr1 cells [28, 29], stimulation with allogeneic APCs or peptide-pulsed APCs in the presence of TGF-β [30-32], ex vivo costimulatory blockade [33] or IFN-γ-conditioned stimulation with alloantigen [34, 35]. In addition, vitamin D or Rapa-conditioned tolerogenic DCs have been used to generate T cells with alloreactive regulatory functions [36-38]. It will be of importance in future investigations which of these strategies is the most superior one. It was also shown that Rapa induces human Treg cells from conventional CD4+ T cells in vitro [39] as well as in vivo [40, 41]. In our experiment, Rapa increased the generation of murine aTreg cells only in combination with aCD4. The ability of TGF-β to induce Treg cells has been known for a long time [42]. Wan and Flavell showed that TGF-β is essential to keep the functionality of CD4+CD25+Foxp3+ in the periphery and that TGF-β has the potential to induce Foxp3 in naïve cells [43].

4-fold higher than that of PAO1 (P = 0 0071) The mutation freque

4-fold higher than that of PAO1 (P = 0.0071). The mutation frequencies of both the 18A and PAO1 Venetoclax cell line biofilm communities were also quantified during biofilm development and dispersal (12 days). The number of morphotypic variants was enumerated to compare the mutation frequency with the frequency of morphotypic variants. The initial mutation frequency for 18A biofilm on day 0 was 3.17 × 10−8 ± 4.87 × 10−8 (Fig. 5a), which was also similar to the mutation frequency of the planktonic culture (3.10 × 10−8 ± 7.53 × 10−9). The mutation frequency decreased during the initial stages of biofilm development to 6.87 × 10−9 ± 7.4 × 10−9 by day 4. On day 8, the mutation frequency increased to 2.65 × 10−8 ± 3.68 × 10−8,

and by day 10, it was 6.11 × 10−8 ± 1.14 × 10−7, similar to MLN0128 concentration the mutation frequency observed at the start of biofilm development and the original planktonic culture. In contrast to PAO1, morphotypic variants appeared in the biofilm of 18A on day 4 and accounted for approximately 49% of the population. On day 10, when the mutation frequency was the highest for strain 18A, approximately 80% of the population consisted of morphotypic variants. Interestingly, by day 12, variants accounted for only 20% of the population at which time the mutation frequency also declined (4.11 × 10−8 ± 3.68 × 10−8). The mutation frequency for the PAO1 biofilm on day 0 was 1.26 × 10−8 ± 9.44 × 10−9 (Fig. 5a), which was similar to the mutation

frequency of the planktonic culture. During the course of biofilm development, it was observed that the mutation frequency decreased from day 0 to day 6 (2.71 × 10−9 ± 1.20 × 10−9

on day 6) and then increased to 5.76 × 10−9 ± 3.21 × 10−9 on day 8 and did not change significantly for the remaining 4 days of the experiment. Morphotypic variants were observed in the biofilms on day 8 and constituted approximately 2% of the total PAO1 biofilm population. The peak number of variants, 12%, was observed on day 10. It was observed that the biofilm of 18A developed more slowly than that of PAO1 (Fig. 5b), Farnesyltransferase which is in accordance with our observation that 18A has a lower growth rate than PAO1 (data not shown). Although the change in mutation frequency of the biofilm community was not statistically significant between the sampling days, there appears to be a positive correlation between the mutation frequency and the variant frequency. For strain 18A, both the mutation frequency and the percentage of variants increased from days 6 to 10 and decreased on day 12. In PAO1, the mutation frequency was observed to increase slightly between days 6–12, which coincided with the emergence of morphotypic variants. Pseudomonas aeruginosa has been shown to establish long-term colonisation of the lungs of CF sufferers. This process of chronic infection has been linked to the appearance of morphotypic variants (e.g. SCVs and mucoid colony types) as well as the selection of variants with reduced overt, or acute, virulence.

5A) Given that C12Id+ germinal centers are not visible prior to

5A). Given that C12Id+ germinal centers are not visible prior to day 7 of infection (Figs. 3A and 4B), this indicated that the presence of helper T cells enhances the extrafollicular-derived C12Id+ Ab responses. Transfer of polyclonal CD4 T cells

also seem to enhance these responses, although these differences did not reach statistical significance (p=0.1; Fig. 5A). Consistent with these findings, frequencies of HA-A/PR8-specific B220lo C12Id+ plasma blasts were higher in TS-1 helper T-cell recipients compared to control mice that did not receive any CD4 T cells (Fig. 5B). Transfer of polyclonal T cells also significantly enhanced the frequencies of the C12Id+ virus-specific cells (Fig. 5B). Whether this is due to the activation of T cells in the isolation process, or non-cognate interaction between Fer-1 B cells and CD4 T cells that could enhance selleck chemicals extrafollicular responses, remains to be studied. Importantly, virus-specific germinal center B-cell frequencies were unaltered by the transfer of specific or non-specific CD4 T cells (Fig. 5C). Thus, the presence of helper CD4 T cells can enhance the magnitude of the extrafollicular B-cell response but cannot shift the quality of the C12Id+ B-cell response toward increased

germinal center formation. Exploiting work by others that previously identified influenza A/PR8 HA-specific Ab of the C12Id as a major component of the early B-cell response to influenza 24, 27, and building on our more recent work identifying influenza HA-specific

B cells by flow cytometry 32, we studied the fate of HA-specific B cells STK38 following influenza virus infection in genetically non-manipulated BALB/c mice. Our studies identify follicular B cells in the regional LN of infected mice as the cell population responsible for much of the early-induced C12Id+ Ab response via their rapid induction of extrafollicular foci. C12Id-expressing B cells also initiated germinal center responses, albeit to a lesser degree and with delayed and irregular kinetics. Increased CD4 T-cell help enhanced the magnitude of the C12-initiated extrafollicular responses. Importantly, it did not shift the response quality toward increased germinal center formation. Together our studies indicate the presence of as yet unknown, presumably innate, signals that cause the expansion but not the initiation of extrafollicular over intrafollicular B-cell responses. Characterization of the early-responding C12Id+ HA-specific B cells failed to provide evidence for a phenotypically distinct B-cell population in the regional LN that could give rise preferentially or exclusively to early Ab-forming foci, as suggested in earlier studies 41.