Acta Paediatr Scand 1986, 75:

Acta Paediatr Scand 1986, 75: Crenolanib order 388–395.CrossRefPubMed 22. Laws E, Vance ML: Radiosurgery for pituitary tumors and craniopharyngiomas. Neurosurgery Clinics of North America 1999, 10: 327–336.PubMed 23. Pan L, Zhang N, Wang E, Wang B, Xu W: Pituitary adenomas: The effect of gamma knife radiosurgery on tumor growth and endocrinopathies. Stereotact Funct Neurosurg 1998, 70: 119–126.CrossRefPubMed 24. Choi JY, Chang JH, Chang JW, Ha Y, Park YG, Chung SS: Radiological and hormonal responses of functioning pituitary adenomas after gamma knife radiosurgery. Yonsei Med J 2003, 44: 602–607.PubMed 25. Kim MS, Lee SI, Sim JH: Gamma Knife radiosurgery for functioning pituitary microadenoma. Stereotact

Funct Neurosurg 1999, 72: 119–124.CrossRefPubMed 26. Becker G, Kocher M, Kortmann RD, Paulsen F, Jeremic B,

Muller RP, Bamberg M: Radiation therapy in the multimodal treatment approach of pituitary adenoma. Strahlenther Onkol 2002, 178: 173–186.CrossRefPubMed 27. Tsang RW, Brierley JD, Panzarella T, Gospodarowicz MK, Sutcliffe SB, Simpson WJ: Role of radiation therapy in clinical hormonally-active pituitary adenomas. Radiother Oncol 1996, 41: 45–53.CrossRefPubMed 28. Salinger DJ, Brady LW, Miyamoto CT: Radiation therapy in the treatment of pituitary adenomas. Am J Clin Oncol 1992, 15: 467–473.CrossRefPubMed 29. McCord MW, Buatti JM, Fennell EM, Mendenhall WM, Marcus RB Jr, Rhoton AL, Grant MB, Friedman WA: Radiotherapy for pituitary www.selleckchem.com/products/Gefitinib.html adenoma: long-term outcome and sequelae. Int J Radiat Oncol Biol Phys 1997, 39: 437–444.CrossRefPubMed 30. Nishioka H, Hirano A, Haraoka J, Nakajima N: Histological changes in the pituitary gland and adenomas following radiotherapy. Neuropathology 2002, 22: 19–25.CrossRefPubMed 31. Post KD, Habas JE: Comparison of long term results between prolactin secreting adenomas and ACTH secreting adenomas. Can J Neurol Sci 1990, 17: 74–77.PubMed 32. Kokubo M, Sasai K, Shibamoto Y, Aoki T, Oya N, Mitsumori M, Takahashi JA, Hashimoto N, Hiraoka M: Long-term results of radiation

therapy for pituitary adenoma. J Neuro oncol 2000, Sinomenine 47: 79–84.CrossRef Competing interests The authors declare that they have no competing interests. Authors’ contributions HW carried out the follow-up of the patients, participated in the irradiation treatment and drafted the manuscript. OC established this gamma knife centre and participated in the irradiation treatment. SBY conceived of the study, and participated in its design and coordination and helped to draft the manuscript. All authors read and approved the final manuscript.”
“Background In spite of the progresses recently registered in the therapy of multiple myeloma (MM), the prognosis for patients affected by this disease remains still poor [1]. MM demonstrate a progressive, usually fatal, course with traditional treatments, generally producing only temporary remissions.

J Exp

Mar Biol Ecol 83:179–193CrossRef Pianka ER (1966) L

J Exp

Mar Biol Ecol 83:179–193CrossRef Pianka ER (1966) Latitudinal gradients in species diversity: a review of concepts. Am Nat 100:33–46CrossRef Reigstad M (2000) Plankton community and vertical flux of biogenic matter in LY2606368 north Norwegian fjords: regulating factors, temporal and spatial variations. Dissertation, Institute of Aquatic Research Environmental Biology, Norwegian College of Fishery Science, Tromsø Reigstad M, Wassmann P (1996) Importance of advection for pelagic-benthic coupling in north Norwegian fjords. Sarsia 80:245–257 Rosenzweig ML (1995) Species diversity in space and time. Cambridge University Press, CambridgeCrossRef Roy K et al (1998) Marine latitudinal diversity gradients: tests of causal hypotheses. Proc Natl Acad Sci 95:3699–3702PubMedCrossRef Salas C, Hergueta E (1986) The molluscan fauna of calcareous concretions of Mesophyllum lichenoides (Ellis) Lemoine. Study of annual cycle diversity. Iberus 6:57–65 Sanders HL (1968) Marine benthic diversity: a comparative study. Am Nat 102:243–282CrossRef Sebens KP (1991) Habitat structure and community dynamics in marine benthic LBH589 ic50 systems. In: Mushinsky HR (ed) Habitat structure: the physical arrangement of objects in space. Chapman and Hall, London Sebens KP, Witting J, Helmuth B (1997) Effects of water flow and branch

spacing on particle capture by the reef coral Madracis mirabilis (Duchassaing and Michelotti). J Exp Mar Biol Ecol 211:1–28CrossRef Senn DG, Glasstetter M (1989) On the occurrence of barnacle -reefs around Cocos-Island, Costa Rica. Senck Marit 20:241–249 Sintes DM (1987) La comunidad de Anélidos Poliquetos de las concreciones de algas calcáreas del litoral catalán. Caracterización de las espicies (In Spanish). Vol 13, Publicaciones del Departamento de Zoología, Universidad de Barcelona, pp 45–54 Sintes DM, Dantart Flavopiridol (Alvocidib) L, Ballesteros M (1987) Moluscos de las concreciones de algas calceras del litoral Catalan (NE España; in Spanish). Lavori 23:445–456 Sjøkartverk N (1957) Den Norske Los (in Norwegian). Norges Sjøkartverk, Oslo, pp 37–44 Sneli J-A (1968) The Lithothamnion community in Nord-Möre, Norway. With notes on the epifauna of Desmarestia viridis

(Müller). Sarsia 31:69–74 Stevens GC (1989) The latitudinal gradient in geographical range: How so many species coexist in the tropics. Am Nat 133:240–256CrossRef Svendsen H (1995) Physical oceanography of coupled fjord-coast systems in northern Norway with special focus on frontal dynamics and tides. In: Leinaas HP (ed) Proceedings from the Mare Nor Symposium on the Ecology of fjords and coastal waters. Elsevier Science, Amsterdam Tande K (1990) Calanus in north Norwegian fjords and in the Barents Sea. In: Sakshaug E, Hopkins CCE, Oeritsland NA (eds) Pro Mare symposium on polar marine ecology. Norwegian Polar Institute Press, Trondheim ten Hove HA (1979) Different causes of mass occurence in Serpulids. In: Rosen BR (ed) Biology and systematics of colonial organisms.

Furthermore, given that anthocyanins also have been described to

Furthermore, given that anthocyanins also have been described to active the nrf-2 transcription factor [20, 48, 49] and induce heat shock proteins [52] it is feasible that blueberry-derived anthocyanins may activate similar and/or parallel adaptive mechanisms within damaged muscle and underlie the findings observed here selleckchem and by others. It is also unclear whether particular anthocyanins or other phytochemicals from fruits (or other sources) are responsible for or synergistic to the benefits reported here. Studies using isolated polyphenolics indicate that they potentially possess diverse

functional efficacy within the body, which may not necessarily complement each other. It is feasible that certain fruit species or even certain cultivars (or combinations thereof) may provide the combination of polyphenolics that synergistically act together to most optimally deliver a specific biological action or actions that complement the adaptive events desired

by exercise training athletes. Conclusions In conclusion, our study provides evidence that ingestion of a New Zealand blueberry AZD6738 beverage prior to and after eccentric muscle damage accelerates recovery of muscle peak isometric strength, independent of the beverages inherent antioxidant properties. Standardizing blueberry fruit intake based on the lean body mass (g/kg), (assuming that the greater the muscle mass, the greater the force produced during the maximal eccentric protocol [53]) may have given more accurate results. This study has practical implications for all who turn to exercise and dietary antioxidant-rich supplements to maintain their health and performance. It is especially of potential relevance to all athletes who compete over successive days as well as to the general sporting community. Although the literature is divided as to the benefits

Liothyronine Sodium of antioxidant supplements in affecting the initial muscle damage/inflammation and subsequent recovery of muscle function, this study supports the idea that blueberry consumption induces cellular adaptive events that serve to accelerate muscle repair and recovery of muscle isometric strength. Identifying specific dietary interventions that complement exercise-induced short-term as well as adaptive responses following various exercise strategies (i.e. aerobic exercise-induced oxidative stress or EIMD) may be of greater importance in maintaining health and athletic performance than the consumption of generic dietary supplements based upon their apparent high antioxidant capacity. Follow up studies are therefore warranted with blueberry as a food to assist exercise and should focus upon dose and timing to ascertain important optimum parameters.

1c) E7AS partially and completely blocked IL-32 and COX-2 expres

1c). E7AS partially and completely blocked IL-32 and COX-2 expression, respectively (Fig. 1c), suggesting that factors other than COX-2 can induce IL-32. It has been reported that a single siRNA targeting the E7 coding region should inhibit the expression of both E6 and E7 proteins simultaneously31 and so E7AS could completely block COX-2 expression. Immunohistochemical staining for both COX-2 and IL-32 revealed the co-localization of these signals in invasive primary cancerous tissues (Fig. 1d). Expressed E7 induced significant increases in the activities of both the IL-32 and COX-2 promoters. As shown in Fig. 2, the HPV-16 E7 oncogene stimulated the promoter activities of click here both IL-32 (Fig. 2a) and COX-2 (Fig. 2b)

in a variety of cervical cancer cell lines, and E7AS specifically neutralized the E7-mediated activation of both the IL-32 (−746/+25) and COX-2 (−880/+9) promoters. In Fig. 2(a), there was no significant increase of IL-32 promoter activity induced by the control IL-32p itself (without E7) in the C33A/pOPI3 control cells (data not shown). Nor was there a significant increase of COX-2 promoter activity induced by the control E7 itself (without COX-2p) in the C33A/pOPI3 cells (Fig. 2b, data not shown). To determine the mechanism underlying the HPV-16 E7-mediated stimulation of COX-2 and IL-32, COX-2 was over-expressed in SiHa and CaSki cells and IL-32

expression was evaluated with RT-PCR and Western blot analyses. The IL-32 mRNA and protein expression levels were increased by COX-2 over-expression (Fig. 3a). In addition, IL-32 and E7 expressions were reduced in a check details dose-dependent manner by treatment with the COX-2-specific inhibitor NS398 in SiHa and CaSki cells (Fig. 3b). The levels of COX-2-derived PGE2 were reduced in the culture media from the NS398-treated SiHa and CaSki cells. Interleukin-32 levels were determined in the supernatants of COX-2 over-expressing and NS398-treated SiHa and CaSki cells using a sandwich IL-32 ELISA as reported Liothyronine Sodium previously,30 and significant expression levels of IL-32 were not detected in the culture media (data not shown) compared with the intracellular expression levels of IL-32. This result

supports the notion that IL-32 would not be secreted from cells as reported previously.12,26 Collectively, these data show that COX-2 is an upstream regulatory factor of HPV-16 E7-mediated IL-32 stimulation. To assess the regulatory effects of IL-32 on the expression of COX-2 mediated by the HPV-16 E7 oncogene in cervical cancer cells, SiHa and CaSki cells were transfected with IL-32γ and IL-32 siRNA, respectively, in independent experiments. The results of RT-PCR and Western blot analyses demonstrated that E7 and COX-2 were down-regulated in cells (over-expressed with IL-32γ) over-expressing IL-32γ and recovered by IL-32 siRNA (Figs 4a and 5a). The broad band of IL-32 proteins detected by Western blotting as shown in Fig. 3(b), suggested the various expressed forms of IL-32 proteins.

For some experiments recombinant mouse IL-10 was added to T-cell

For some experiments recombinant mouse IL-10 was added to T-cell cultures (1 ng/ml; eBioscience). Proliferation was assessed by pulsing cultures overnight with 0·5 μCi/well of [3H]thymidine overnight and performing scintillation counts. Culture supernatants were harvested daily over 4 days. Expression levels of interferon-γ, IL-2, IL-4, IL-5, IL-10, IL-17 and tumour necrosis LDK378 datasheet factor-α in supernatant samples were quantified by means of a cytofluorimetry-based ELISA system according to the manufacturer’s instructions (Flowcytomix; Bender Medsystem GmbH, Vienna, Austria). Cells were suspended in

FACS buffer (3% fetal calf serum, 5 mm EDTA in PBS). Cells were incubated with conjugated monoclonal antibodies in the presence of Fc blockers (clone 2.4G2). All data acquisition was performed on a FACSCalibur flow cytometer (Becton-Dickinson, San Jose, CA). The anti-mouse monoclonal antibodies used (Becton-Dickinson) were: CD4-FITC, CD44-phycoerythrin, CD62L-peridinin chlorophyll protein complex, CD25-allophycocyanin and Foxp3-phycoerythrin. T cells selleck chemicals were identified as CD3+ and either CD4+ CD8− for CD4 T cells or CD4− CD8+ for CD8+ T cells. CD44, CD62L and CD25 expression was used to assess

T-cell activation status. For FACS, regulatory T (Treg) cells were characterized as CD4+ CD44intermediate/high CD25+ cells.12 All values were expressed as the mean ± standard error of the mean (SEM). Statistical analysis was calculated by the two-tailed unpaired t-test using graphpad prism software (GraphPad Software, La Jolla, CA). A P-value < 0·05 was considered statistically significant. To confirm that the proliferation inhibition observed among ASC−/− CD3+ T cells in response to anti-CD3/CD28 stimulation9 is specifically linked to ASC deficiency and so not a consequence of a general NALP3 inflammasome dysfunction, we initially compared the proliferative response

of ASC−/− and NALP3−/− CD3+ T cells. When compared with ASC−/− CD3+ T cells, NALP3−/− CD3+ T cells did not display an impaired proliferative to response to anti-CD3/CD28 stimulation (Fig. 1a), suggesting that this ASC-associated T-cell defect is NALP3 inflammasome-independent. We next investigated whether this ASC−/− T-cell phenomenon is restricted to a specific T-cell subset or if it affects T cells more globally. Therefore purified CD4+ and CD8+ T cells from ASC+/+ and ASC−/− mice were stimulated separately with plate-bound anti-CD3/CD28 and their proliferation was assessed over time. When compared with similarly stimulated WT controls, ASC−/− CD4+ (Fig. 1b) and CD8+ (Fig. 1c) T cells displayed no impairment in their proliferative response upon activation. Furthermore, no alteration in the regulation of T-cell activation markers (CD44, CD62L and CD25) was observed on ASC−/− CD4+ (Fig. 1d) and CD8+ (Fig. 1e) T cells following activation compared with WT controls.

Non-specific binding was blocked using 10% goat serum in TBST (0·

Non-specific binding was blocked using 10% goat serum in TBST (0·1 m Tris–HCl, pH 7·5; 0·15 m NaCl; 0·1% Tween-20) for 30 min. Sections were then incubated for 60 min with the following primary antibodies: CD3e-biotin, CD11b, CD11c-allophycocyanin (APC), CD103-phycoerythrin

(PE), CD11c-biotin (BD Biosciences, Stockholm, Sweden) and with IgD (Biolegend, San Diego, CA), diluted in TBST. Unlabelled RXDX-106 datasheet antibodies were detected using Cy5-conjugated anti-rat IgG (Jackson ImmunoResearch, West Grove, PA), and biotinylated antibodies were detected using fluorophore tyramide (PerkinElmer, Waltham, MA). Tissue sections were mounted in Vectashield with DAPI (Vector Laboratories, Burlingame, CA), and analysed using laser scanning confocal microscopy (Leica TSP-2; Leica, Heidelberg, Germany). Images were analysed using leica lcs software (Leica, San Jose, CA) and Adobe Photoshop CS3. Intracellular staining for Foxp3 was carried out using a Mouse Regulatory T Cell Staining kit (eBioscience, San Diego, CA). 7-Amino-actinomycin D (7AAD) was used to exclude dead cells. The following conjugated antibodies were used for surface staining: CD3e-APC, CD4-Alexa-700, CD8a-PE-Cy7, CD11b-APC-Cy7,

CD11c-Pacific blue, CD45R-Pacific blue, CD45R-Alexa Fluor 488, MHC-II-Alexa-700, Saracatinib supplier KJ1-26-PE and Foxp3-PE (eBioscience), CD19-APC, CD25-APC-Cy7, CD62L-APC, CD103-PE (BD Bioscience), and streptavidin-Qdot 605 (Invitrogen). CD172a antibody was provided by Dr Karl Lagenaur and biotinylated in-house. Flow cytometry was performed on an LSR:II (BD Bioscience) and results were analysed using flowjo software (Tree Star, Ashland, OR). CD4+ T cells were enriched from spleens and LN of DO11.10 mice by positive selection magnetic separation using a MACS LS-column (Miltenyi Biotec, BergischGladbach, Germany). CD4+ cells were stained with 2·5 μm 5,6-carboxyfluorescein diacetate succinimidyl ester (CFSE; Invitrogen) and 2·5 × 106 to 5 × 106 cells were Meloxicam injected intravenously into recipient CD47−/− and WT mice. The following day, mice were fed 10 mg OVA (grade V; Sigma, Stockholm, Sweden) in the presence or absence of 10 μg CT (Sigma) in 3% NaHCO3, or injected with 100 μg OVA intravenously. After 3 days, organs

were harvested and CD4+ T-cell proliferation was analysed by CFSE profiling. CD47−/− and WT mice were fed PBS or OVA (5 or 50 mg). Ten days later, all mice were challenged subcutaneously with 100 μg OVA in incomplete Freund’s adjuvant (IFA). Draining LN (inguinal) were harvested 1 week later and cells were re-stimulated with low-endotoxin OVA. Three days later, [3H]thymidine was added for 6 hr, then cells were harvested, and thymidine incorporation was measured using a β-counter. The stimulation index was defined as cellular proliferation in the OVA-fed group in relation to the PBS-fed group normalized to 0%. Wild-type mice that received PBS were used as reference for OVA-fed WT mice, and PBS-fed CD47−/− mice were reference for OVA-fed CD47−/− mice.

, 1999) Imiquimod at 0 5 μg mL−1 was optimal for human PBMC prod

, 1999). Imiquimod at 0.5 μg mL−1 was optimal for human PBMC production of TNF-α, IFN-γ, IL-1, IL-6, IL-8, IL-10, IL-12, GM-CSF, G-CSF, and MIP-1α, with a 24-h incubation (Stanley, 2002). Although we PD-0332991 ic50 did not define in the present

study as to which cells in murine PBMC elaborate the cytokines we identified, other studies, with imiquimod, have indicated that the cells in human PBMC producing proinflammatory cytokines are monocyte/macrophages and B cells (Megyeri et al., 1995). Analysis of cellular requirements in human PBMC for cytokine production induced by imiquimod indicated that T-lymphocytes were responsible for IFN-γ production, but required IL-12 and IFN-γ from imiquimod-stimulated macrophages (Wagner et al., 1999). Other studies with TLR-7 agonists suggest that monocytes are the main cells found in abundance in human peripheral blood that are responsive. This was also true of the stronger response induced by TLR-8 and TLR-7/8 agonists, as would be relevant to 3M-003 (Gorden et al., 2005). Although responses of mouse spleen ALK phosphorylation cells to imiquimod

have been reported (Wagner et al., 1999), we are not aware of studies using mouse PBMC and imiquimod. Here, we report novel findings that 3M-003-stimulated mouse PBMC produce high levels of TNF-α and IL-12, but little to no IFN-γ in the time frame examined. Supernatants from mouse PBMC cultures containing high levels of TNF-α and IL-12 were sufficient to induce enhanced candidacidal activity in macrophages, neutrophils, and monocytes. That macrophages are upregulated by PBMC-produced factors in supernatants was evidenced by the 3M-003 carryover in supernatants being much less than the concentrations we show required for consistent direct macrophage activation. Supernatant neutralization and/or addition (e.g. TNF-α, IL-12, or TNF-α+IL-12) experiments are warranted to further elucidate the phagocyte activation mechanism induced by supernatants. These compounds are potentially useful for antifungal therapy.

This could especially be important in the common entity, neonatal candidiasis (Chapman & Faix, 2003), because TLR-8 agonists appear to be particularly potent activators of the neonatal immune system (Philbin & Levy, 2007). It would be of interest to ascertain whether the antifungal activity would extend to hyphal forms and to other fungi. Systemic use of these Amrubicin compounds is under study as an antineoplastic (Dudek et al., 2007; Harrison et al., 2007; Smith et al., 2007). Cytokine induction has been noted after oral administration (Dahl, 2002; Harandi et al., 2003). An additional possible mechanism of action of the imidazoquinolines is TLR-independent immunomodulation by antagonism of adenosine receptors (Philbin & Levy, 2007). Agonists of human TLR-8 can also reverse the function of regulatory T cells; caution may need to be exercised for possible overabundance of an inflammatory response with such agents (Philbin & Levy, 2007).

According to the developers’ instructions, the possible scores fo

According to the developers’ instructions, the possible scores for each domain ranged from 0 (best health) to 100 (worst health).8 All data are expressed as the mean ± standard deviation (SD) or frequency and percentage. The internal consistency reliability (Cronbach’s alpha) of the IPSS and KHQ was calculated for all domains except the single-item domains. A Cronbach’s alpha coefficient greater than 0.80 is considered excellent, while a value greater than 0.70 is acceptable.16 Exploratory factor analysis (principal component analysis) with

varimax rotation, which means the construct validity, was used to explore the underlying factor structure of the KHQ. The criteria used to indicate the appropriateness of factor analysis were a significant Bartlett’s test of sphericity and a approved range of values of Kaiser–Meyer–Olkin BVD-523 purchase (KMO, 0.7 to 1.0). Factors were extracted based on the Kaiser’s criterion of eigenvalues greater than 1. Furthermore, the discriminant validity of the KHQ was assessed using one-way analysis of variance (ANOVA) tests with post hoc tests (Games-Howell

method) by comparing the subscales in the KHQ domains between mild, moderate, and severe LUTS group. The total, filling, and voiding IPSS between the three LUTS groups were also compared. All data were analyzed using SPSS version Pritelivir mw 17.0 (SPSS Inc., Chicago, IL, USA). A P-value (-)-p-Bromotetramisole Oxalate of 0.05 was considered statistically significant. Among 393 men with at least one point in the IPSS, about 7.9% (n = 31) of participants had severe LUTS, while 25.4% (n = 100) had moderate LUTS, and 66.7% (n = 262) had mild LUTS. The mean ages for severe, moderate, and mild LUTS groups were 65.4 ± 11.1, 66.1 ± 11.5, and 60.9 ± 11.6 years, respectively. Table 1 shows the descriptive statistics and internal consistency reliability of the IPSS and the KHQ. The Cronbach’s α coefficients for eight KHQ subscales ranged from 0.750 to 0.943, while the Cronbach’s

α coefficient was 0.889, 0.714, and 0.889 for total, filling, and voiding IPSS, respectively. The appropriateness of factor analysis is supported by Bartlett’s test (χ2 = 5167.6, P < 0.001) and the KMO measure of sampling adequacy (KMO = 0.858). Table 2 shows that three factors were identified, and totally explained about 70.0% of the variance, while the explained variance for factors 1, 2, and 3 were 30.9, 23.4, and 15.3%, respectively. Table 3 shows the mean scores in the IPSS and the KHQ subscales by three LUTS groups. The results indicated that there were significant differences in mean scores for the total, filling, and voiding IPSS between severe, moderate, and mild groups (all P < 0.001).

Similar to the Helicobacter model, IL-23 was responsible for indu

Similar to the Helicobacter model, IL-23 was responsible for inducing IL-17 production and colon-specific VX-809 research buy tissue inflammation, and depletion of the Sca-1+ ILCs prevented development of colitis [3]. The idea that IL-17 production by ILCs can contribute to autoimmune disease has also been explored in humans. IL-17-producing cells are increased in the intestine of patients with ulcerative colitis and Crohn’s disease [8]. CD3− cells contributed significantly to the production of IL-17, both IL-17a and IL-17f mRNA

transcripts were increased in CD3− cells isolated from the intestines of patients with IBD as compared with transcripts in healthy controls [8]. In addition, there is an increased frequency of ILCs in the colon and ileum of patients Rapamycin mw with Crohn’s disease but not ulcerative

colitis [8]. However, since the absolute numbers of IL-17-producing ILCs in the inflamed intestine are very small, it is still unclear whether these cells play a direct role in driving IBD. Therefore, further studies are needed to determine their exact role. There have been a small number of reports showing that NK cells produce IL-17. Since human NKR-LTi cells have been shown to secrete IL-17 [82], careful analysis and interpretation of the results are essential to avoid confusion between IL-17 production by NKR-LTi cells and that by classical NK cells. In the steady state, NK cells in the spleen do not express RORγt [5]; however, upon infection with Toxoplasma gondii, splenic NK cells have enhanced

RORγt expression and secrete IL-17 [4]. A recent report has also shown that CD56+CCR4+ human peripheral blood NK cells produce both IL-17 and IFN-γ and express the transcription factors RORγt and Tbet [98]. These cells are not NKR-LTi cells, since the NK cells in this study did not express IL-7R (CD127), nor IL-23R, and since NKR-LTi cells are not thought to exist in human peripheral blood [82, 89]. iNKT cells are a subset of T cells that express a semi-invariant TCR that recognizes glycolipids presented by CD1d molecules expressed on APCs. There have been a number of recent reports demonstrating that iNKT Olopatadine cells play a role in host protection against infection via the production of IL-17. Expression of RORγt in developing iNKT precursor cells is associated with the development of a preprogrammed IL-17-producing subset that does not express NK1.1 [99]. The signals that induce RORγt expression in iNKT precursors and lineage commitment have not yet been defined. These NK1.1− iNKT cells are capable of secreting IL-17 not only in response to stimulation with the synthetic ligand α-galactosylceramide or its analogue PBS-57, but also following stimulation with natural ligands, including LPS or glycolipids derived from Sphingomonas wittichii and Borrelia burgdorferi [100]. This IL-17-producing NK1.1− subset is present at high frequency in the lung, comprising up to 40% of pulmonary iNKT cells in naïve mice.